101
|
Yamaguchi M, Murata T, Ramos JW. The overexpressed regucalcin represses the growth via regulating diverse pathways linked to EGF signaling in human ovarian cancer SK-OV-3 cells: Involvement of extracellular regucalcin. Life Sci 2023; 314:121328. [PMID: 36584916 DOI: 10.1016/j.lfs.2022.121328] [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: 10/18/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
AIMS Regucalcin, which plays a multifunctional role in cell regulation, contributes as a suppressor in carcinogenesis. Survival of cancer patients is prolonged with high expression of regucalcin in tumor tissues. Ovarian cancer is the most lethal in gynecologic malignancies. This study elucidates the repressive role of regucalcin on the growth of human ovarian cancer SK-OV-3 cells that are resistant to cytotoxic cancer drugs. MATERIALS AND METHODS SK-OV-3 wild type-cells and regucalcin-overexpressing cells (transfectants) were cultured in Dulbecco's Modification of Eagle's Medium containing 10 % fetal bovine serum. KEY FINDINGS Colony formation and proliferation of SK-OV-3 cells were repressed by regucalcin overexpression. The suppressive effects of regucalcin on proliferation were independent of cell death. The proliferation of SK-OV-3 wild-type cells was repressed by various inhibitors, including cell cycle, signaling processes, and transcriptional activity. The effects of all inhibitors were not revealed in transfectants, suggesting the involvement of multiple signaling pathways in regucalcin effects. Of note, the overexpressed regucalcin declined the levels of Ras, Akt, mitogen-activating protein kinase, NF-κB p65, β-catenin, and STAT3, while it raised the levels of tumor suppressors p53 and Rb, and cell cycle inhibitor p21. Interestingly, the stimulatory effects of epidermal growth factor (EGF) on cell proliferation were blocked in regucalcin-overexpressing cells. Extracellular regucalcin repressed the proliferation independent of the death of SK-OV-3 cells and blocked EGF-enhanced cell proliferation. SIGNIFICANCES The overexpressed regucalcin may repress cell proliferation by targeting diverse signal pathways, including EGF signaling. This study offers a novel approach to the treatment of ovarian cancer with regucalcin.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, HI 96813, USA.
| | - Tomiyasu Murata
- Laboratory of Molecular Biology, Faculty of Pharmacy, Meijo University, Yagotoyama 150, Tempaku, Nagoya 468-8503, Japan
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, HI 96813, USA
| |
Collapse
|
102
|
Wallis B, Bowman KR, Lu P, Lim CS. The Challenges and Prospects of p53-Based Therapies in Ovarian Cancer. Biomolecules 2023; 13:159. [PMID: 36671544 PMCID: PMC9855757 DOI: 10.3390/biom13010159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
It has been well established that mutations in the tumor suppressor gene, p53, occur readily in a vast majority of cancer tumors, including ovarian cancer. Typically diagnosed in stages three or four, ovarian cancer is the fifth leading cause of death in women, despite accounting for only 2.5% of all female malignancies. The overall 5-year survival rate for ovarian cancer is around 47%; however, this drops to an abysmal 29% for the most common type of ovarian cancer, high-grade serous ovarian carcinoma (HGSOC). HGSOC has upwards of 96% of cases expressing mutations in p53. Therefore, wild-type (WT) p53 and p53-based therapies have been explored as treatment options via a plethora of drug delivery vehicles including nanoparticles, viruses, polymers, and liposomes. However, previous p53 therapeutics have faced many challenges, which have resulted in their limited translational success to date. This review highlights a selection of these historical p53-targeted therapeutics for ovarian cancer, why they failed, and what the future could hold for a new generation of this class of therapies.
Collapse
Affiliation(s)
| | | | | | - Carol S. Lim
- Department of Molecular Pharmaceutics, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
103
|
Chang YH, Wu KC, Harnod T, Ding DC. Comparison of the Cost and Effect of Combined Conditioned Medium and Conventional Medium for Fallopian Tube Organoid Cultures. Cell Transplant 2023; 32:9636897231160216. [PMID: 36919683 PMCID: PMC10021093 DOI: 10.1177/09636897231160216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Fallopian tube epithelial cells (FTEC) are thought to be the cell of origin of high-grade serous ovarian carcinoma. FTEC organoids can be used as research models for the disease. Nevertheless, culturing organoids requires a medium supplemented with several expensive growth factors. We proposed that a combined conditioned medium based on the composition of the fallopian tubes, including epithelial, stromal, and endothelial cells could enhance FTEC organoid formation. We derived two primary culture cell lines from the fimbria portion of the fallopian tubes. The organoids were split into conventional or combined medium groups based on what medium they were grown in and compared. The number and size of the organoids were evaluated. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) were used to evaluate gene and protein expression (PAX8, FOXJ1, beta-catenin, and stemness genes). Enzyme-linked immunosorbent assay was used to measure Wnt3a and RSPO1 in both mediums. DKK1 and LiCl were added to the mediums to evaluate their influence on beta-catenin signaling. The growth factor in the combined medium was evaluated by the growth factor array. We found that the conventional medium was better for organoids regarding proliferation (number and size). In addition, WNT3A and RSPO1 concentrations were too low in the combined medium and needed to be added making the cost equivalent to the conventional medium. However, the organoid formation rate was 100% in both groups. Furthermore, the combined medium group had higher PAX8 and stemness gene expression (OLFM4, SSEA4, LGR5, B3GALT5) when compared with the conventional medium group. Wnt signaling was evident in the organoids grown in the conventional medium but not in the combined medium. PLGF, IGFBP6, VEGF, bFGF, and SCFR were found to be enriched in the combined medium. In conclusion, the combined medium could successfully culture organoids and enhance PAX8 and stemness gene expression. However, the conventional medium was a better medium for organoid proliferation. The expense of both mediums was comparable. The benefit of using a combined medium requires further exploration.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,Tzu Chi University, Hualien
| | - Kun-Chi Wu
- Tzu Chi University, Hualien.,Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Tomor Harnod
- Tzu Chi University, Hualien.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Dah-Ching Ding
- Tzu Chi University, Hualien.,Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,Institute of Medical Sciences, Collagen of Medicine, Tzu Chi University, Hualien
| |
Collapse
|
104
|
Wang Y, Huang P, Wang BG, Murdock T, Cope L, Hsu FC, Wang TL, Shih IM. Spatial Transcriptomic Analysis of Ovarian Cancer Precursors Reveals Reactivation of IGFBP2 during Pathogenesis. Cancer Res 2022; 82:4528-4541. [PMID: 36206311 PMCID: PMC9808976 DOI: 10.1158/0008-5472.can-22-1620] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/15/2022] [Accepted: 10/03/2022] [Indexed: 01/24/2023]
Abstract
Elucidating the earliest pathogenic steps in cancer development is fundamental to improving its early detection and prevention. Ovarian high-grade serous carcinoma (HGSC), a highly aggressive cancer, mostly originates from the fallopian tube epithelium through a precursor stage, serous tubal intraepithelial carcinoma (STIC). In this study, we performed spatial transcriptomic analysis to compare STICs, carcinoma, and their matched normal fallopian tube epithelium. Several differentially expressed genes in STICs and carcinomas were involved in cancer metabolism and detected in a larger independent transcriptomic dataset of ovarian HGSCs. Among these, insulin-like growth factor binding protein-2 (IGFBP2) was found to undergo DNA hypomethylation and to be increased at the protein level in STICs. Pyrosequencing revealed an association of IGFBP2 expression with the methylation state of its proximal enhancer, and 5-azacytidine treatment increased IGFBP2 expression. In postmenopausal fallopian tubes, where most STICs are detected, IGFBP2 immunoreactivity was detected in all 38 proliferatively active STICs but was undetectable in morphologically normal tubal epithelia, including those with TP53 mutations. In premenopausal fallopian tubes, IGFBP2 expression was limited to the secretory epithelium at the proliferative phase, and estradiol treatment increased IGFBP2 expression levels. IGFBP2 knockdown suppressed the growth of IGFBP2-expressing tubal epithelial cells via inactivation of the AKT pathway. Taken together, demethylation of the proximal enhancer of IGFBP2 drives tumor development by maintaining the increased IGFBP2 required for proliferation in an otherwise estrogen-deprived, proliferation-quiescent, and postmenopausal tubal microenvironment. SIGNIFICANCE Molecular studies of the earliest precursor lesions of ovarian cancer reveal a role of IGFBP2 in propelling tumor initiation, providing new insights into ovarian cancer development.
Collapse
Affiliation(s)
- Yeh Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peng Huang
- Biostatistics Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brant G. Wang
- Department of Pathology, Inova Fairfax Hospital, Falls Church, Virginia
| | - Tricia Murdock
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leslie Cope
- Biostatistics Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fang-Chi Hsu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland,Department of Gynecology and Obstetrics and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland,Department of Gynecology and Obstetrics and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
105
|
Cassani C, Rossi C, Camnasio CA, Urtis M, Fiandrino G, Grasso M, Zanellini F, Lucioni M, D’Ambrosio G, Di Toro A, Rossi M, Roccio M, Ferrari A, Secondino S, Nappi RE, Arbustini E, Paulli M, Spinillo A, Cesari S. Pathologic Findings at Risk Reducing Surgery in BRCA and Non- BRCA Mutation Carriers: A Single-Center Experience. Diagnostics (Basel) 2022; 12:diagnostics12123054. [PMID: 36553061 PMCID: PMC9776991 DOI: 10.3390/diagnostics12123054] [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: 11/17/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Risk-reducing surgery (RRS) is recommended in BRCA-mutated carriers because of their increased risk of developing ovarian cancer, while its role is still discussed for women harboring mutations in non-BRCA homologous repair genes. The aim of this study was to retrospectively evaluate the occurrence of pathological findings in a high-risk population undergoing RRS in San Matteo Hospital, Pavia between 2012 and 2022, and correlate their genetic and clinical outcomes, comparing them with a control group. The final cohort of 190 patients included 85 BRCA1, 63 BRCA2, 11 CHEK2, 7 PALB2, 4 ATM, 1 ERCC5, 1 RAD51C, 1 CDH1, 1 MEN1, 1 MLH1 gene mutation carriers and 15 patients with no known mutation but with strong familial risk. Occult invasive serous carcinoma (HGSC) and serous tubal intraepithelial carcinoma (STIC) were diagnosed in 12 (6.3%) women, all of them BRCA carriers. No neoplastic lesion was diagnosed in the non-BRCA group, in women with familial risk, or in the control group. Oral contraceptive use and age ≤45 at surgery were both found to be favorable factors. While p53 signature and serous tubal intraepithelial lesion (STIL) were also seen in the control group and in non-BRCA carriers, STIC and HGSC were only found in BRCA1/2 mutation carriers.
Collapse
Affiliation(s)
- Chiara Cassani
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Unit of Obstetrics and Gynecology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Chiara Rossi
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Cristina Angela Camnasio
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Unit of Obstetrics and Gynecology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Mario Urtis
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Giacomo Fiandrino
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Maurizia Grasso
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Francesca Zanellini
- Unit of Obstetrics and Gynecology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Marco Lucioni
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Gioacchino D’Ambrosio
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Alessandro Di Toro
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Margherita Rossi
- Unit of Obstetrics and Gynecology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Marianna Roccio
- Unit of Obstetrics and Gynecology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Alberta Ferrari
- General Surgery III—Breast Surgery, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Simona Secondino
- Unit of Medical Oncology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Rossella Elena Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Eloisa Arbustini
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Marco Paulli
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Unit of Obstetrics and Gynecology, University of Pavia, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Stefania Cesari
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
- Correspondence:
| |
Collapse
|
106
|
Kamal IM, Temerik DF, Yassin EH, Mosad E, A H, Hussien MT. Prognostic Outcome of Mesenchymal Transition Biomarkers in Correlation with EGFR Expression in Epithelial Ovarian Carcinoma Patients. Asian Pac J Cancer Prev 2022; 23:4213-4225. [PMID: 36580004 PMCID: PMC9971466 DOI: 10.31557/apjcp.2022.23.12.4213] [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: 07/24/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND CD44 is an epithelial-mesenchymal transition (EMT) surface receptor that regulates the interactivity between the cells and the extracellular matrix, thereby promoting cell migration. The epidermal growth factor receptor (EGFR) family is a trans-membrane kinase-related protein. It regulates cell adhesion proteins, which may promote cell proliferation and invasiveness. Mesenchymal epithelial transition (MET) is another EMT receptor that stimulates cell proliferation, invasion, survival, and angiogenesis. This study aimed to evaluate the prognostic impact of CD44, EGFR expressions, and MET gene amplification in epithelial ovarian cancer (EOC). METHODS This is a retrospective cohort study, including 85 cases of EOC. CD44 and EGFR expressions were evaluated in both epithelial and stromal cells by immunohistochemistry. Tumor cells also underwent a cytogenetic analysis using fluorescent in situ hybridization (FISH) to detect MET gene amplification. RESULTS High CD44 expression in tumors was significantly associated with serous subtypes (P=0.001), peritoneal deposits (P=0.002), and advanced stage (P=0.002). EGFR high tumor expression demonstrated a significant association with lymph node metastasis (P=0.038) and the advanced stage of EOC (P=0.016). Increased copy number of the MET gene was significantly associated with partial therapy response (P=0.030). CD44 and EGFR tumor high expression was associated with poor overall survival (OS). In addition, MET gene gain in tumors was associated with a shorter OS (P=0.000). CONCLUSION EMT biomarkers (CD44 and MET) and EGFR expression in EOC are independent prognostic factors for OS. MET gene increase copy number was detected in cases of serous neoplasm and associated with poor survival and minimal therapy response.
Collapse
Affiliation(s)
- Israa Mostafa Kamal
- Department of Oncologic Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt.
| | - Doaa F Temerik
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt.
| | - Etemad H Yassin
- Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Eman Mosad
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt.
| | - Hanan A
- Department of Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut Egypt.
| | - Marwa T Hussien
- Department of Oncologic Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt. ,For Correspondence:
| |
Collapse
|
107
|
Husby A, Wohlfahrt J, Melbye M. Pregnancy duration and ovarian cancer risk: A 50-year nationwide cohort study. Int J Cancer 2022; 151:1717-1725. [PMID: 35751416 PMCID: PMC9796914 DOI: 10.1002/ijc.34192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 01/07/2023]
Abstract
A woman's reproductive history is strongly associated with her risk of ovarian cancer. However, it is unclear how pregnancies of different duration impact a woman's ovarian cancer risk, and therefore, what part of a pregnancy explains the protective effect. Using a cohort of all Danish women followed from 1968 to 2018, with prospectively registered information on reproductive history (eg, gestational duration of pregnancies, tubal ligation and resection and hormonal pharmaceutical use), we investigated the effect of pregnancy duration on ovarian cancer risk. We adjusted for potential confounders, such as age at pregnancy and time since pregnancy, using log-linear Poisson regression to isolate the effect of pregnancy duration on ovarian cancer risk. Among 2.5 million Danish women with 4.4 million pregnancies, a pregnancy was associated with a reduction of ovarian cancer risk of 21% (95% CI, 14%-28%), 26% (95% CI, 21%-31%), 12% (95% CI, 7%-17%) and 3% (95% CI, -5% to 11%) compared to one less, for the first, second, third and fourth pregnancy, respectively (P < .001 for heterogeneity), with similar effects of induced abortions, spontaneous abortions and childbirths. Sensitivity analysis of age at pregnancy, time since pregnancy and other potential confounders did not change these findings. The reduced ovarian cancer risk associated with pregnancy is primarily driven by the first three pregnancies, with similar effects of induced abortion, spontaneous abortions and childbirth, suggesting that mainly exposure to early pregnancy factors, and not pregnancy duration, protect against ovarian cancer.
Collapse
Affiliation(s)
- Anders Husby
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK,Department of Epidemiology ResearchStatens Serum InstitutCopenhagenDenmark
| | - Jan Wohlfahrt
- Department of Epidemiology ResearchStatens Serum InstitutCopenhagenDenmark
| | - Mads Melbye
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark,Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA,Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway,K.G. Jebsen Center for Genetic EpidemiologyNorwegian University of Science and TechnologyTrondheimNorway
| |
Collapse
|
108
|
Lin Y, Zhou X, Ni Y, Zhao X, Liang X. Metabolic reprogramming of the tumor immune microenvironment in ovarian cancer: A novel orientation for immunotherapy. Front Immunol 2022; 13:1030831. [PMID: 36311734 PMCID: PMC9613923 DOI: 10.3389/fimmu.2022.1030831] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic tumor, with the highest mortality rate. Numerous studies have been conducted on the treatment of ovarian cancer in the hopes of improving therapeutic outcomes. Immune cells have been revealed to play a dual function in the development of ovarian cancer, acting as both tumor promoters and tumor suppressors. Increasingly, the tumor immune microenvironment (TIME) has been proposed and confirmed to play a unique role in tumor development and treatment by altering immunosuppressive and cytotoxic responses in the vicinity of tumor cells through metabolic reprogramming. Furthermore, studies of immunometabolism have provided new insights into the understanding of the TIME. Targeting or activating metabolic processes of the TIME has the potential to be an antitumor therapy modality. In this review, we summarize the composition of the TIME of ovarian cancer and its metabolic reprogramming, its relationship with drug resistance in ovarian cancer, and recent research advances in immunotherapy.
Collapse
|
109
|
Ghezelayagh TS, Kohrn BF, Fredrickson J, Manhardt E, Radke MR, Katz R, Gray HJ, Urban RR, Pennington KP, Liao JB, Doll KM, Simons EJ, Burzawa JK, Goff BA, Speiser P, Swisher EM, Norquist BM, Risques RA. Uterine lavage identifies cancer mutations and increased TP53 somatic mutation burden in individuals with ovarian cancer. CANCER RESEARCH COMMUNICATIONS 2022; 2:1282-1292. [PMID: 36311816 PMCID: PMC9615025 DOI: 10.1158/2767-9764.crc-22-0314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Current screening methods for ovarian cancer (OC) have failed to demonstrate a significant reduction in mortality. Uterine lavage combined with TP53 ultra-deep sequencing for the detection of disseminated OC cells has emerged as a promising tool, but this approach has not been tested for early-stage disease or non-serous histologies. In addition, lavages carry multiple background mutations, the significance of which is poorly understood. Uterine lavage was collected preoperatively in 34 patients undergoing surgery for suspected ovarian malignancy including 14 patients with benign disease and 20 patients with OC (6 non-serous and 14 high grade serous-like (serous)). Ultra-deep duplex sequencing (~3000x) with a panel of common OC genes identified the tumor mutation in 33% of non-serous (all early stage) and in 79% of serous cancers (including four early stage). In addition, all lavages carried multiple somatic mutations (average of 25 mutations per lavage), more than half of which corresponded to common cancer driver mutations. Driver mutations in KRAS, PIK3CA, PTEN, PPP2R1A and ARID1A presented as larger clones than non-driver mutations and with similar frequency in lavages from patients with and without OC, indicating prevalent somatic evolution in all patients. Driver TP53 mutations, however, presented as significantly larger clones and with higher frequency in lavages from individuals with OC, suggesting that TP53-specific clonal expansions are linked to ovarian cancer development. Our results demonstrate that lavages capture cancer cells, even from early-stage cancers, as well as other clonal expansions and support further exploration of TP53 mutation burden as a potential OC risk factor.
Collapse
Affiliation(s)
- Talayeh S. Ghezelayagh
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Brendan F. Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Jeanne Fredrickson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Enna Manhardt
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Marc R. Radke
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Heidi J. Gray
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Renata R. Urban
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Kathryn P. Pennington
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - John B. Liao
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Kemi M. Doll
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Elise J. Simons
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Jennifer K. Burzawa
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Barbara A. Goff
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | | | - Elizabeth M. Swisher
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Barbara M. Norquist
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Rosa Ana Risques
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
- Corresponding Author: Rosa Ana Risques, Laboratory Medicine and Pathology, University of Washington, HSB, E506, Seattle, WA 98195. Phone: 206-616-4976; E-mail:
| |
Collapse
|
110
|
Enroth S, Ivansson E, Lindberg JH, Lycke M, Bergman J, Reneland A, Stålberg K, Sundfeldt K, Gyllensten U. Data-driven analysis of a validated risk score for ovarian cancer identifies clinically distinct patterns during follow-up and treatment. COMMUNICATIONS MEDICINE 2022; 2:124. [PMID: 36196264 PMCID: PMC9526736 DOI: 10.1038/s43856-022-00193-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 09/23/2022] [Indexed: 11/05/2022] Open
Abstract
Background Ovarian cancer is the eighth most common cancer among women and due to late detection prognosis is poor with an overall 5-year survival of 30-50%. Novel biomarkers are needed to reduce diagnostic surgery and enable detection of early-stage cancer by population screening. We have previously developed a risk score based on an 11-biomarker plasma protein assay to distinguish benign tumors (cysts) from malignant ovarian cancer in women with adnexal ovarian mass. Methods Protein concentrations of 11 proteins were characterized in plasma from 1120 clinical samples with a custom version of the proximity extension assay. The performance of the assay was evaluated in terms of prediction accuracy based on receiver operating characteristics (ROC) and multiple hypothesis adjusted Fisher's Exact tests on achieved sensitivity and specificity. Results The assay's performance is validated in two independent clinical cohorts with a sensitivity of 0.83/0.91 and specificity of 0.88/0.92. We also show that the risk score follows the clinical development and is reduced upon treatment, and increased with relapse and cancer progression. Data-driven modeling of the risk score patterns during a 2-year follow-up after diagnosis identifies four separate risk score trajectories linked to clinical development and survival. A Cox proportional hazard regression analysis of 5-year survival shows that at time of diagnosis the risk score is the second-strongest predictive variable for survival after tumor stage, whereas MUCIN-16 (CA-125) alone is not significantly predictive. Conclusion The robust performance of the biomarker assay across clinical cohorts and the correlation with clinical development indicates its usefulness both in the diagnostic work-up of women with adnexal ovarian mass and for predicting their clinical course.
Collapse
Affiliation(s)
- Stefan Enroth
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, SE-75108 Uppsala, Sweden ,grid.462826.c0000 0004 5373 8869Swedish Collegium for Advanced Study, Thunbergsvägen 2, SE-752 38 Uppsala, Sweden
| | - Emma Ivansson
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, SE-75108 Uppsala, Sweden
| | - Julia Hedlund Lindberg
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, SE-75108 Uppsala, Sweden
| | - Maria Lycke
- grid.8761.80000 0000 9919 9582Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at Gothenburg University, SE-416 85 Gothenburg, Sweden
| | | | | | - Karin Stålberg
- grid.8993.b0000 0004 1936 9457Department of Women’s and Children’s Health, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Karin Sundfeldt
- grid.8761.80000 0000 9919 9582Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at Gothenburg University, SE-416 85 Gothenburg, Sweden
| | - Ulf Gyllensten
- grid.8993.b0000 0004 1936 9457Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, SE-75108 Uppsala, Sweden
| |
Collapse
|
111
|
A Revised Markov Model Evaluating Oophorectomy at the Time of Hysterectomy for Benign Indication: Age 65 Years Revisited. Obstet Gynecol 2022; 140:520-521. [PMID: 36356241 DOI: 10.1097/aog.0000000000004912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
112
|
Konishi I, Abiko K, Hayashi T, Yamanoi K, Murakami R, Yamaguchi K, Hamanishi J, Baba T, Matsumura N, Mandai M. Peritoneal dissemination of high-grade serous ovarian cancer: pivotal roles of chromosomal instability and epigenetic dynamics. J Gynecol Oncol 2022; 33:e83. [PMID: 36032027 PMCID: PMC9428305 DOI: 10.3802/jgo.2022.33.e83] [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: 06/02/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Epithelial ovarian cancer remains the lethal gynecological malignancy in women. The representative histotype is high-grade serous carcinoma (HGSC), and most patients with HGSC present at advanced stages with peritoneal dissemination. Since the peritoneal dissemination is the most important factor for poor prognosis of the patients, complete exploration for its molecular mechanisms is mandatory. In this narrative review, being based on the clinical, pathologic, and genomic findings of HGSC, chromosomal instability and epigenetic dynamics have been discussed as the potential drivers for cancer development in the fallopian tube, acquisition of cancer stem cell (CSC)-like properties, and peritoneal metastasis of HGSC. The natural history of carcinogenesis with clonal evolution, and adaptation to microenvironment of peritoneal dissemination of HGSC should be targeted in the novel development of strategies for prevention, early detection, and precision treatment for patients with HGSC.
Collapse
Affiliation(s)
- Ikuo Konishi
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takuma Hayashi
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
113
|
Effect of ovulation IGF and HGF signaling on the oncogenesis of murine epithelial ovarian cancer cell ID8. Exp Cell Res 2022; 419:113323. [PMID: 36030968 DOI: 10.1016/j.yexcr.2022.113323] [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: 05/06/2022] [Revised: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022]
Abstract
The incidence and mortality of epithelial ovarian cancer (EOC) are increasing in Taiwan and worldwide. The prognosis of this disease has improved little in the last few decades due to insufficient knowledge of the etiology. Previous studies on the role of ovulation in the development of EOC have unveiled IGF2, HGF, and other carcinogens in ovulatory follicular fluid (FF) that exert transformation activities on the exposed fallopian tube fimbria epithelium. However, an orthotopic proof in an animal model is lacking. By using the murine ID8 EOC cells and the syngenic transplantation model, this study explored the effect of FF on the oncogenesis of mouse ovarian cancer. We found FF promoted clonogenicity and anchorage-independent growth of ID8 cells, largely through the IGF-1R and cMET signaling. In contrast, FF modestly promoted cell proliferation independent of the two signals and did not affect cell migration and invasion. Transplantation of ID8 cells into the ovarian bursa of C57BL6/J mice orthotopically grew ovarian tumors and metastasized to the peritoneum with ascites formation. The tumorigenic rate and severity of the disease were positively correlated with the level of IGF-1R and cMET receptors on the cell surface. Our data demonstrated that ovulation, through the signaling of IGF/IGF-1R and HGF/cMET, promotes oncogenic phenotypes in a murine EOC model. The results provide further proof of the carcinogenic effect of ovulation in the development of EOC.
Collapse
|
114
|
Multiomics characterization implicates PTK7 in ovarian cancer EMT and cell plasticity and offers strategies for therapeutic intervention. Cell Death Dis 2022; 13:714. [PMID: 35977930 PMCID: PMC9386025 DOI: 10.1038/s41419-022-05161-5] [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: 02/27/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 01/21/2023]
Abstract
Most patients with ovarian cancer (OC) are diagnosed at a late stage when there are very few therapeutic options and a poor prognosis. This is due to the lack of clearly defined underlying mechanisms or an oncogenic addiction that can be targeted pharmacologically, unlike other types of cancer. Here, we identified protein tyrosine kinase 7 (PTK7) as a potential new therapeutic target in OC following a multiomics approach using genetic and pharmacological interventions. We performed proteomics analyses upon PTK7 knockdown in OC cells and identified novel downstream effectors such as synuclein-γ (SNCG), SALL2, and PP1γ, and these findings were corroborated in ex vivo primary samples using PTK7 monoclonal antibody cofetuzumab. Our phosphoproteomics analyses demonstrated that PTK7 modulates cell adhesion and Rho-GTPase signaling to sustain epithelial-mesenchymal transition (EMT) and cell plasticity, which was confirmed by high-content image analysis of 3D models. Furthermore, using high-throughput drug sensitivity testing (525 drugs) we show that targeting PTK7 exhibited synergistic activity with chemotherapeutic agent paclitaxel, CHK1/2 inhibitor prexasertib, and PLK1 inhibitor GSK461364, among others, in OC cells and ex vivo primary samples. Taken together, our study provides unique insight into the function of PTK7, which helps to define its role in mediating aberrant Wnt signaling in ovarian cancer.
Collapse
|
115
|
Vang R, Shih IM. Serous tubal intraepithelial carcinoma: What Do We Really Know at this Point? Histopathology 2022; 81:542-555. [PMID: 35859323 DOI: 10.1111/his.14722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/18/2022] [Accepted: 06/23/2022] [Indexed: 11/27/2022]
Abstract
Serous tubal intraepithelial carcinoma (STIC) is the earliest morphologically recognizable step in the development of invasive high-grade serous carcinoma of the fallopian tube. Lesions occurring prior to STIC within the carcinogenic sequence for the pathogenesis of invasive high-grade serous carcinoma include the p53 signature and secretory cell outgrowth (SCOUT). Variable histologic criteria have been used for diagnosing STIC, but a combination of morphology and immunohistochemistry for p53/Ki-67 improves interobserver agreement. Half of all carcinomas identified in risk-reducing salpingo-oophorectomy specimens are in the form of STIC; however, STIC also may be incidentally found on occasion in specimens from women at low or average risk of ovarian/tubal/peritoneal carcinoma. TP53 mutation is the earliest known DNA sequence alteration in STIC and almost all invasive high-grade serous carcinomas of the ovary and peritoneum. Data on the clinical behavior of STIC are limited. While the short-term follow-up in the prior literature suggests a low risk of malignant progression, a more recent meta-analysis indicates a 10-year risk of 28%. STIC probably should be best regarded as a lesion with uncertain malignant potential at present, and future molecular analysis will help classify those with higher risk of dissemination. This review article provides an update on the current knowledge of STIC and related issues.
Collapse
Affiliation(s)
- Russell Vang
- Departments of Pathology (Division of Gynecologic Pathology), The Johns Hopkins University School of Medicine; Baltimore, MD, USA.,Gynecology & Obstetrics, The Johns Hopkins University School of Medicine; Baltimore, MD, USA
| | - Ie-Ming Shih
- Departments of Pathology (Division of Gynecologic Pathology), The Johns Hopkins University School of Medicine; Baltimore, MD, USA.,Gynecology & Obstetrics, The Johns Hopkins University School of Medicine; Baltimore, MD, USA.,Oncology, The Johns Hopkins University School of Medicine; Baltimore, MD, USA
| |
Collapse
|
116
|
Xu H, Tang Y, Liu L, Yan J, Qin L. Downregulation of lncRNA ASMTL-AS1 in Epithelial Ovarian Cancer Correlates with Worse Prognosis and Cancer Progression. Horm Metab Res 2022; 54:481-488. [PMID: 35835145 DOI: 10.1055/a-1872-0546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Given the characters of "Silent killer", epithelial ovarian cancer (EOC) usually suffered late diagnosis and poor prognosis. Therefore, this study aimed to explore the prognostic significance of ASMTL-AS1 in EOC and investigated the effect of lncRNA ASMTL-AS1 dysregulation on tumor cellular function. ASMTL-AS1 expression was analyzed in 133 EOC tissues and five kinds of cell lines by RT-qPCR. The expression of ASMTL-AS1 was tested for correlation with clinical data using the chi-square test and clinical follow-up using Kaplan-Meier method with log-rank test. Further, the prognostic parameters in predicting EOC overall survival were assessed by using multivariate Cox proportional hazards analysis. In vitro assays, including MTT assay and transwell assay, were conducted using EOC cell lines with overexpression of ASMTL-AS1. In tumorous tissues and cell lines, ASMTL-AS1 was lowly expressed compared with normal ones. This downregulation was associated with the advanced FIGO stage, positive ascites cytology, and lymph node. In particular, low levels of ASMTL-AS1 were revealed to have a high prognostic impact on EOC. ASMTL-AS1 overexpression strongly decreased cell proliferation, migration, and invasion in vitro partly by moderating miR-1228-3p. This study demonstrates a significant role for lowly expressed ASMTL-AS1 in EOC allowing for the prediction of prognosis for EOC. Considering that ASMTL-AS1 is strongly involved in cell growth and invasion, ASMTL-AS1 may be a promising marker for EOC prognosis and therapy.
Collapse
Affiliation(s)
- Hui Xu
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Yan Tang
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Lu Liu
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Jie Yan
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Li Qin
- Department of Obstetrics and Gynecology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| |
Collapse
|
117
|
Differential epithelial and stromal LGR5 expression in ovarian carcinogenesis. Sci Rep 2022; 12:11200. [PMID: 35778589 PMCID: PMC9249864 DOI: 10.1038/s41598-022-15234-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/21/2022] [Indexed: 11/08/2022] Open
Abstract
Lgr5 has been identified as a marker of the stem/progenitor cells in the murine ovary and oviduct by lineage tracing. However, little is known regarding LGR5 expression or its functional significance in human ovary tissues. Here, using RNA in situ hybridization and/or immunohistochemistry, we thoroughly investigated LGR5 expression in normal human ovaries, fallopian tubes and various ovarian tumors. We discovered that LGR5 expression is negligible in the human ovary surface epithelium, whereas ovarian stromal cells normally express low levels of LGR5. Remarkably, fallopian tube epithelium, inclusion cysts and serous cystadenomas with a Müllerian phenotype expressed high levels of LGR5, and LGR5 expression was restricted to PAX8+/FOXJ1- secretory cells of the tubal epithelium. Strong stromal LGR5 expression without epithelial LGR5 expression was consistently observed in the path from serous cystadenoma to serous borderline tumor to low grade serous carcinoma (LGSC). Unlike LGSC, high grade serous carcinoma (HGSC), clear cell carcinoma, endometrioid carcinomas displayed various epithelial-stromal LGR5 expression. Notably, high levels of LGR5 expression were observed in serous tubal intraepithelial carcinoma, which slightly declined in invasive HGSC. LGR5 expression was significantly associated with improved progression-free survival in HGSC patients. Moreover, in vitro assays demonstrated that LGR5 expression suppressed tumor proliferation and migratory capabilities. Taken together, these findings indicate a tumor-suppressive role for LGR5 in the progression of HGSC.
Collapse
|
118
|
Peremiquel-Trillas P, Paytubi S, Pelegrina B, Frias-Gomez J, Carmona Á, Martínez JM, de Francisco J, Benavente Y, Barahona M, Briansó F, Canet-Hermida J, Caño V, Vidal A, Zanca A, Baixeras N, Rodríguez A, Fernández-Gonzalez S, Dueñas N, Càrdenas L, Aytés Á, Bianchi I, Pavón MÀ, Reventós J, Capellà G, Gómez D, Diaz M, Ponce J, Brunet J, Matias-Guiu X, Bosch FX, de Sanjosé S, Alemany L, Pineda M, Marin F, Costas L. An Integrated Approach for the Early Detection of Endometrial and Ovarian Cancers (Screenwide Study): Rationale, Study Design and Pilot Study. J Pers Med 2022; 12:jpm12071074. [PMID: 35887570 PMCID: PMC9324683 DOI: 10.3390/jpm12071074] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Screenwide is a case-control study (2017−2021) including women with incident endometrial and ovarian cancers (EC and OC), BRCA1/2 and MMR pathogenic variant carriers, and age-matched controls from three centers in Spain. Participants completed a personal interview on their sociodemographic factors, occupational exposure, medication, lifestyle, and medical history. We collected biological specimens, including blood samples, self-collected vaginal specimens, cervical pap-brush samples, uterine specimens, and, when available, tumor samples. The planned analyses included evaluation of the potential risk factors for EC/OC; evaluation of molecular biomarkers in minimally invasive samples; evaluation of the cost-effectiveness of molecular tests; and the generation of predictive scores to integrate different epidemiologic, clinical, and molecular factors. Overall, 182 EC, 69 OC, 98 BRCA pathogenic variant carriers, 104 MMR pathogenic variant carriers, and 385 controls were enrolled. The overall participation rate was 85.7%. The pilot study using 61 samples from nine EC cases and four controls showed that genetic variants at the variant allele fraction > 5% found in tumors (n = 61 variants across the nine tumors) were detected in paired endometrial aspirates, clinician-collected cervical samples, and vaginal self-samples with detection rates of 90% (55/61), 79% (48/61), and 72% (44/61) by duplex sequencing, respectively. Among the controls, only one somatic mutation was detected in a cervical sample. We enrolled more than 800 women to evaluate new early detection strategies. The preliminary data suggest that our methodological approach could be useful for the early detection of gynecological cancers.
Collapse
Affiliation(s)
- Paula Peremiquel-Trillas
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Sonia Paytubi
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
| | - Beatriz Pelegrina
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
| | - Jon Frias-Gomez
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Álvaro Carmona
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
| | - José Manuel Martínez
- Department of Gynecology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.M.M.); (M.B.); (S.F.-G.); (J.P.)
| | - Javier de Francisco
- Department of Anesthesiology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.d.F.); (V.C.)
| | - Yolanda Benavente
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Marc Barahona
- Department of Gynecology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.M.M.); (M.B.); (S.F.-G.); (J.P.)
| | - Ferran Briansó
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, 08028 Barcelona, Spain;
- Roche Diagnostics, Sant Cugat del Vallès, 08174 Barcelona, Spain
| | - Júlia Canet-Hermida
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
| | - Víctor Caño
- Department of Anesthesiology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.d.F.); (V.C.)
| | - August Vidal
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
- Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain; (A.Z.); (N.B.); (A.R.)
| | - Alba Zanca
- Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain; (A.Z.); (N.B.); (A.R.)
| | - Núria Baixeras
- Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain; (A.Z.); (N.B.); (A.R.)
| | - Axel Rodríguez
- Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain; (A.Z.); (N.B.); (A.R.)
| | - Sergi Fernández-Gonzalez
- Department of Gynecology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.M.M.); (M.B.); (S.F.-G.); (J.P.)
| | - Núria Dueñas
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
| | - Laura Càrdenas
- Department of Gynecology and Obstetrics, Hospital Universitari Dr. Josep Trueta, 17007 Girona, Spain;
| | - Álvaro Aytés
- Program against Cancer Therapeutic Resistance (ProCURE), IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Ilaria Bianchi
- ASSIR Delta, Serveis d’Atenció Primària Delta del Llobregat, Direcció d’Atenció Primària Costa de Ponent, Gerència Territorial Metropolitana Sud, Institut Català de la Salut, 08006 Barcelona, Spain;
| | - Miquel Àngel Pavón
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Jaume Reventós
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, 08017 Barcelona, Spain;
| | - Gabriel Capellà
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
| | - David Gómez
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
| | - Mireia Diaz
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Jordi Ponce
- Department of Gynecology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain; (J.M.M.); (M.B.); (S.F.-G.); (J.P.)
| | - Joan Brunet
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
- Medical Oncology Department, Catalan Institute of Oncology, Doctor Josep Trueta Girona University Hospital, 17007 Girona, Spain
| | - Xavier Matias-Guiu
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
- Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain; (A.Z.); (N.B.); (A.R.)
| | - Francesc Xavier Bosch
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
- Universitat Oberta de Catalunya, 08018 Barcelona, Spain
| | - Silvia de Sanjosé
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
- Consultant, National Cancer Institute, Bethesda, MA 20814, USA
| | - Laia Alemany
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
| | - Marta Pineda
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
| | - Fátima Marin
- Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain; (J.C.-H.); (N.D.); (G.C.); (J.B.); (M.P.); (F.M.)
- Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain; (A.V.); (X.M.-G.)
| | - Laura Costas
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain; (P.P.-T.); (S.P.); (B.P.); (J.F.-G.); (Á.C.); (Y.B.); (M.À.P.); (D.G.); (M.D.); (F.X.B.); (L.A.)
- Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain;
- Correspondence:
| |
Collapse
|
119
|
Liberto JM, Chen SY, Shih IM, Wang TH, Wang TL, Pisanic TR. Current and Emerging Methods for Ovarian Cancer Screening and Diagnostics: A Comprehensive Review. Cancers (Basel) 2022; 14:2885. [PMID: 35740550 PMCID: PMC9221480 DOI: 10.3390/cancers14122885] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
With a 5-year survival rate of less than 50%, ovarian high-grade serous carcinoma (HGSC) is one of the most highly aggressive gynecological malignancies affecting women today. The high mortality rate of HGSC is largely attributable to delays in diagnosis, as most patients remain undiagnosed until the late stages of -disease. There are currently no recommended screening tests for ovarian cancer and there thus remains an urgent need for new diagnostic methods, particularly those that can detect the disease at early stages when clinical intervention remains effective. While diagnostics for ovarian cancer share many of the same technical hurdles as for other cancer types, the low prevalence of the disease in the general population, coupled with a notable lack of sensitive and specific biomarkers, have made the development of a clinically useful screening strategy particularly challenging. Here, we present a detailed review of the overall landscape of ovarian cancer diagnostics, with emphasis on emerging methods that employ novel protein, genetic, epigenetic and imaging-based biomarkers and/or advanced diagnostic technologies for the noninvasive detection of HGSC, particularly in women at high risk due to germline mutations such as BRCA1/2. Lastly, we discuss the translational potential of these approaches for achieving a clinically implementable solution for screening and diagnostics of early-stage ovarian cancer as a means of ultimately improving patient outcomes in both the general and high-risk populations.
Collapse
Affiliation(s)
- Juliane M. Liberto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (J.M.L.); (I.-M.S.); (T.-L.W.)
| | - Sheng-Yin Chen
- School of Medicine, Chang Gung University, 33302 Taoyuan, Taiwan;
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (J.M.L.); (I.-M.S.); (T.-L.W.)
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Tza-Huei Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (J.M.L.); (I.-M.S.); (T.-L.W.)
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
120
|
Hsu CF, Seenan V, Wang LY, Chu TY. Ovulation Enhances Intraperitoneal and Ovarian Seedings of High-Grade Serous Carcinoma Cells Originating from the Fallopian Tube: Confirmation in a Bursa-Free Mouse Xenograft Model. Int J Mol Sci 2022; 23:ijms23116211. [PMID: 35682896 PMCID: PMC9181345 DOI: 10.3390/ijms23116211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 01/25/2023] Open
Abstract
Background: Recently, new paradigms for the etiology and origin of ovarian high-grade serous carcinoma (HGSC) have emerged. The carcinogens released during ovulation transform fallopian tube epithelial cells, exfoliating and metastasizing to the peritoneal organs, including the ovaries. Solid in vivo evidence of the paradigms in a mouse model is urgently needed but is hampered by the differing tubo-ovarian structures. In mice, there is a bursa structure surrounding the distal oviduct and ovary. This, on one hand, prevents the direct influence of ovulatory follicular fluid (FF) on the exfoliated tumor cells. On the other hand, it hinders the seeding of exfoliated tumor cells into the ovary. Methods: In this study, we created a bursa-free mouse xenograft model to examine the effect of superovulation on peritoneal and ovarian metastases of transformed human tubal epithelial cells after intraperitoneal injection in NSG mice. Results: The bursa-free mouse model showed a better effect of ovulation on peritoneal metastasis. In this model, superovulation increased the number of transformed human tubal epithelial cell seedlings after intraperitoneal injection. Compared to the bursa-intact state, bursa-free ovaries were more vulnerable to external tumor seeding in either normal ovulation or superovulation state. Conclusions: This study provides the first in vivo evidence that intraperitoneal spreading of tubal HGSC cells is enhanced by ovulation. This study also demonstrated a mouse model for studying ovary-peritoneum interaction in cancer development.
Collapse
Affiliation(s)
- Che-Fang Hsu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (C.-F.H.); (V.S.); (L.-Y.W.)
| | - Vaishnavi Seenan
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (C.-F.H.); (V.S.); (L.-Y.W.)
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| | - Liang-Yuan Wang
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (C.-F.H.); (V.S.); (L.-Y.W.)
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970, Taiwan
| | - Tang-Yuan Chu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (C.-F.H.); (V.S.); (L.-Y.W.)
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970, Taiwan
- Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Life Sciences, Tzu Chi University, Hualien 970, Taiwan
- Correspondence:
| |
Collapse
|
121
|
Rashid S, Arafah MA, Akhtar M. The Many Faces of Serous Neoplasms and Related Lesions of the Female Pelvis: A Review. Adv Anat Pathol 2022; 29:154-167. [PMID: 35180738 PMCID: PMC8989637 DOI: 10.1097/pap.0000000000000334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ovarian serous tumors and related lesions are one of the most common conditions of the female genital tract. While ovarian high-grade serous carcinoma carries high mortality and adverse prognosis, most other serous lesions have better clinical behavior. In recent years, significant progress has been made in understanding the nature and histogenesis of these lesions that has contributed to better and more precise clinical management. Most of the high-grade serous carcinomas involve the ovaries and/or peritoneum, although in most cases, their origin seems to be in the fallopian tube. This view is supported by the recognition of precursor lesions in the fallopian tube, such as p53 signature and serous tubular in situ carcinoma. This paper presents salient morphologic, immunohistochemical, and molecular data related to serous tumors and related lesions of the female pelvis and discusses the histogenetic interrelationship among these lesions in light of current knowledge.
Collapse
Affiliation(s)
- Sameera Rashid
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, Qatar
| | - Maria A. Arafah
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Akhtar
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
122
|
Aging accelerates while multiparity delays tumorigenesis in mouse models of high-grade serous carcinoma. Gynecol Oncol 2022; 165:552-559. [DOI: 10.1016/j.ygyno.2022.03.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
|
123
|
Risk-Reducing Options for High-Grade Serous Gynecologic Malignancy in BRCA1/2. Curr Oncol 2022; 29:2132-2140. [PMID: 35323372 PMCID: PMC8946924 DOI: 10.3390/curroncol29030172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/25/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer (OC) is the leading cause of death among women with gynecologic malignancy. Breast Cancer Susceptibility Gene 1 (BRCA 1) and Breast Cancer Susceptibility Gene 2 (BRCA 2) germline mutations confer an estimated 20 to 40 times increased risk of OC when compared to the general population. The majority of BRCA-associated OC is identified in the late stage, and no effective screening method has been proven to reduce mortality. Several pharmacologic and surgical options exist for risk-reduction of gynecologic malignancy in BRCA 1/2 mutation carriers. This review summarizes up-to-date research on pharmacologic risk-reducing interventions, including the oral contraceptive pill, acetylsalicylic acid/nonsteroidal anti inflammatory drugs (ASA/NSAID) therapy, and denosumab, and surgical risk-reducing interventions, including risk-reducing bilateral salpingo-oophorectomy, salpingectomy with delayed oophorectomy, and hysterectomy at the time of risk-reducing bilateral salpingo-oophorectomy.
Collapse
|
124
|
Chen YC, Yang CW, Chan TF, Farooqi AA, Chang HS, Yen CH, Huang MY, Chang HW. Cryptocaryone Promotes ROS-Dependent Antiproliferation and Apoptosis in Ovarian Cancer Cells. Cells 2022; 11:cells11040641. [PMID: 35203294 PMCID: PMC8870566 DOI: 10.3390/cells11040641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
Cryptocaryone (CPC) is a bioactive dihydrochalcone derived from Cryptocarya plants, and its antiproliferation was rarely reported, especially for ovarian cancer (OVCA). This study aimed to examine the regulation ability and mechanism of CPC on three histotypes of OVCA cells (SKOV3, TOV-21G, and TOV-112D). In a 24 h MTS assay, CPC showed antiproliferation effects to OVCA cells, i.e., IC50 values 1.5, 3, and 9.5 μM for TOV-21G, SKOV3, and TOV-112D cells. TOV-21G and SKOV3 cells showed hypersensitivity to CPC when applied for exposure time and concentration experiments. For biological processes, CPC stimulated the generation of reactive oxygen species and mitochondrial superoxide and promoted mitochondrial membrane potential dysfunction in TOV-21G and SKOV3 cells. Apoptosis was detected in OVCA cells through subG1 accumulation and annexin V staining. Apoptosis signaling such as caspase 3/7 activities, cleaved poly (ADP-ribose) polymerase, and caspase 3 expressions were upregulated by CPC. Specifically, the intrinsic and extrinsic apoptotic caspase 9 and caspase 8 were overexpressed in OVCA cells following CPC treatment. Moreover, CPC also stimulated DNA damages in terms of γH2AX expression and increased γH2AX foci. CPC also induced 8-hydroxy-2’-deoxyguanosine DNA damages. These CPC-associated principal biological processes were validated to be oxidative stress-dependent by N-acetylcysteine. In conclusion, CPC is a potential anti-OVCA natural product showing oxidative stress-dependent antiproliferation, apoptosis, and DNA damaging functions.
Collapse
Affiliation(s)
- Yu-Chieh Chen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-C.C.); (T.-F.C.)
| | - Che-Wei Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-W.Y.); (H.-S.C.); (C.-H.Y.)
| | - Te-Fu Chan
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-C.C.); (T.-F.C.)
- Department of Obstetrics and Gynecology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan;
| | - Hsun-Shuo Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-W.Y.); (H.-S.C.); (C.-H.Y.)
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-W.Y.); (H.-S.C.); (C.-H.Y.)
| | - Ming-Yii Huang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (M.-Y.H.); (H.-W.C.); Tel.: +886-7-312-1101 (ext. 7158) (M.-Y.H. & H.-W.C.)
| | - Hsueh-Wei Chang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (M.-Y.H.); (H.-W.C.); Tel.: +886-7-312-1101 (ext. 7158) (M.-Y.H. & H.-W.C.)
| |
Collapse
|
125
|
Oleanolic Acid (OA) Targeting UNC5B Inhibits Proliferation and EMT of Ovarian Cancer Cell and Increases Chemotherapy Sensitivity of Niraparib. JOURNAL OF ONCOLOGY 2022; 2022:5887671. [PMID: 35035481 PMCID: PMC8758276 DOI: 10.1155/2022/5887671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022]
Abstract
Objective To investigate the effect of OA on proliferation, migration, and epithelial-mesenchymal transition (EMT) of ovarian cancer cells by inhibiting UNC5B and to study its mechanism. Methods TCGA database was used to analyze the expression of UNC5B in ovarian cancer and its relationship with prognosis. The expression of UNC5B in ovarian cancer cells was detected by qPCR assay. qRT-PCR was used to detect the changes of EMT markers after different treatments. CCK-8 assay was used to detect cell proliferation, transwell assay was used to evaluate cell migration, and clonogenesis assay was used to evaluate the effect of UNC5B on ovarian cancer cell proliferation. Meanwhile, the synergistic effect of OA on niraparib was evaluated. Results UNC5B was highly expressed in ovarian cancer, and its expression was negatively correlated with the prognosis of ovarian cancer patients. UNC5B was highly expressed in ovarian cancer cells SKOV3 and OVCA420 compared with normal ovarian epithelial cells. In addition, silencing UNC5B inhibits the proliferation, invasion, clonogenesis, and EMT processes of ovarian cancer cells. OA inhibits proliferation, invasion, and clonogenesis of ovarian cancer cells by inhibiting UNC5B and increases the antitumor activity of niraparib. Conclusion UNC5B acts as an oncogenic gene in ovarian cancer. OA inhibits ovarian cancer cell proliferation, migration, and EMT by targeting UNC5B and increases the antitumor effect of niraparib. UNC5B is expected to be a new potential therapeutic target for ovarian cancer. OA may be used as an antitumor drug and deserves further study.
Collapse
|
126
|
Akahane T, Masuda K, Hirasawa A, Kobayashi Y, Ueki A, Kawaida M, Misu K, Nakamura K, Nagai S, Chiyoda T, Yamagami W, Hayashi S, Kataoka F, Banno K, Sugano K, Okita H, Kosaki K, Nishihara H, Aoki D. TP53 variants in p53 signatures and the clonality of STICs in RRSO samples. J Gynecol Oncol 2022; 33:e50. [PMID: 35557033 PMCID: PMC9250861 DOI: 10.3802/jgo.2022.33.e50] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tomoko Akahane
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Akira Hirasawa
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
- Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Arisa Ueki
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Miho Kawaida
- Department of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Kumiko Misu
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Shimpei Nagai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Shigenori Hayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Fumio Kataoka
- Department of Obstetrics and Gynecology, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kokichi Sugano
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
- Department of genetic medicine, Kyoundo Hospital, Sasaki Foundation, Tokyo, Japan
| | - Hajime Okita
- Department of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
127
|
Ritch SJ, Telleria CM. The Transcoelomic Ecosystem and Epithelial Ovarian Cancer Dissemination. Front Endocrinol (Lausanne) 2022; 13:886533. [PMID: 35574025 PMCID: PMC9096207 DOI: 10.3389/fendo.2022.886533] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is considered the deadliest gynecological disease and is normally diagnosed at late stages, at which point metastasis has already occurred. Throughout disease progression, EOC will encounter various ecosystems and the communication between cancer cells and these microenvironments will promote the survival and dissemination of EOC. The primary tumor is thought to develop within the ovaries or the fallopian tubes, both of which provide a microenvironment with high risk of causing DNA damage and enhanced proliferation. EOC disseminates by direct extension from the primary tumors, as single cells or multicellular aggregates. Under the influence of cellular and non-cellular factors, EOC spheroids use the natural flow of peritoneal fluid to reach distant organs within the peritoneal cavity. These cells can then implant and seed distant organs or tissues, which develop rapidly into secondary tumor nodules. The peritoneal tissue and the omentum are two common sites of EOC metastasis, providing a microenvironment that supports EOC invasion and survival. Current treatment for EOC involves debulking surgery followed by platinum-taxane combination chemotherapy; however, most patients will relapse with a chemoresistant disease with tumors developed within the peritoneum. Therefore, understanding the role of the unique microenvironments that promote EOC transcoelomic dissemination is important in improving patient outcomes from this disease. In this review article, we address the process of ovarian cancer cellular fate at the site of its origin in the secretory cells of the fallopian tube or in the ovarian surface epithelial cells, their detachment process, how the cells survive in the peritoneal fluid avoiding cell death triggers, and how cancer- associated cells help them in the process. Finally, we report the mechanisms used by the ovarian cancer cells to adhere and migrate through the mesothelial monolayer lining the peritoneum. We also discuss the involvement of the transcoelomic ecosystem on the development of chemoresistance of EOC.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada
- *Correspondence: Carlos M. Telleria, ; orcid.org/0000-0003-1070-3538
| |
Collapse
|
128
|
Nelfinavir Induces Cytotoxicity towards High-Grade Serous Ovarian Cancer Cells, Involving Induction of the Unfolded Protein Response, Modulation of Protein Synthesis, DNA Damage, Lysosomal Impairment, and Potentiation of Toxicity Caused by Proteasome Inhibition. Cancers (Basel) 2021; 14:cancers14010099. [PMID: 35008264 PMCID: PMC8750028 DOI: 10.3390/cancers14010099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC) accounts for 70% of all ovarian-cancer-related deaths. Mainstay treatment with platinum-based drugs following surgery results in favorable outcomes in the majority of patients; however, in >80% of cases, the disease relapses with eventual drug resistance. As such, urgent development of improved alternative therapies is necessary for HGSOC patients with lower life expectancy. Rapid repurposing of market available drugs for cancer therapy is a cost-effective alternative to bypass the decade-long traditional drug development pipeline. Among potential drug-repurposing candidates, nelfinavir (NFV)—an anti-infective agent to treat acquired immunodeficiency syndrome (AIDS)—has shown anti-cancer effects against diverse cancers; however, its remedial benefits against HGSOC are unknown. In this study, we explored how NFV targets HGSOC cells obtained from patients at platinum-sensitive and -resistant stages. We observed beneficial efficacy elicited by NFV against HGSOC in both disease conditions through multiple mechanistic avenues, suggesting positive drug-repurposing prospects. Abstract High-grade serous ovarian cancer (HGSOC) is a significant cause of mortality among women worldwide. Traditional treatment consists of platinum-based therapy; however, rapid development of platinum resistance contributes to lower life expectancy, warranting newer therapies to supplement the current platinum-based protocol. Repurposing market-available drugs as cancer therapeutics is a cost- and time-effective way to avail new therapies to drug-resistant patients. The anti-HIV agent nelfinavir (NFV) has shown promising toxicity against various cancers; however, its role against HGSOC is unknown. Here, we studied the effect of NFV against HGSOC cells obtained from patients along disease progression and carrying different sensitivities to platinum. NFV triggered, independently of platinum sensitivity, a dose-dependent reduction in the HGSOC cell number and viability, and a parallel increase in hypo-diploid DNA content. Moreover, a dose-dependent reduction in clonogenic survival of cells escaping the acute toxicity was indicative of long-term residual damage. In addition, dose- and time-dependent phosphorylation of H2AX indicated NFV-mediated DNA damage, which was associated with decreased survival and proliferation signals driven by the AKT and ERK pathways. NFV also mediated a dose-dependent increase in endoplasmic reticulum stress-related molecules associated with long-term inhibition of protein synthesis and concurrent cell death; such events were accompanied by a proapoptotic environment, signaled by increased phospho-eIF2α, ATF4, and CHOP, increased Bax/Bcl-2 ratio, and cleaved executer caspase-7. Finally, we show that NFV potentiates the short-term cell cycle arrest and long-term toxicity caused by the proteasome inhibitor bortezomib. Overall, our in vitro study demonstrates that NFV can therapeutically target HGSOC cells of differential platinum sensitivities via several mechanisms, suggesting its prospective repurposing benefit considering its good safety profile.
Collapse
|
129
|
Barr JL, Kruse A, Restaino AC, Tulina N, Stuckelberger S, Vermeer SJ, Williamson CS, Vermeer DW, Madeo M, Stamp J, Bell M, Morgan M, Yoon JY, Mitchell MA, Budina A, Omran DK, Schwartz LE, Drapkin R, Vermeer PD. Intra-Tumoral Nerve-Tracing in a Novel Syngeneic Model of High-Grade Serous Ovarian Carcinoma. Cells 2021; 10:3491. [PMID: 34944001 PMCID: PMC8699855 DOI: 10.3390/cells10123491] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Dense tumor innervation is associated with enhanced cancer progression and poor prognosis. We observed innervation in breast, prostate, pancreatic, lung, liver, ovarian, and colon cancers. Defining innervation in high-grade serous ovarian carcinoma (HGSOC) was a focus since sensory innervation was observed whereas the normal tissue contains predominantly sympathetic input. The origin, specific nerve type, and the mechanisms promoting innervation and driving nerve-cancer cell communications in ovarian cancer remain largely unknown. The technique of neuro-tracing enhances the study of tumor innervation by offering a means for identification and mapping of nerve sources that may directly and indirectly affect the tumor microenvironment. Here, we establish a murine model of HGSOC and utilize image-guided microinjections of retrograde neuro-tracer to label tumor-infiltrating peripheral neurons, mapping their source and circuitry. We show that regional sensory neurons innervate HGSOC tumors. Interestingly, the axons within the tumor trace back to local dorsal root ganglia as well as jugular-nodose ganglia. Further manipulations of these tumor projecting neurons may define the neuronal contributions in tumor growth, invasion, metastasis, and responses to therapeutics.
Collapse
Affiliation(s)
- Jeffrey L. Barr
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Allison Kruse
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Anthony C. Restaino
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
- Sanford School of Medicine, University of South Dakota, 414 East Clark St., Vermillion, SD 57069, USA
| | - Natalia Tulina
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Sarah Stuckelberger
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Samuel J. Vermeer
- Lincoln High School, 2900 South Cliff Avenue, Sioux Falls, SD 57105, USA;
| | - Caitlin S. Williamson
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Daniel W. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Marianna Madeo
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Jillian Stamp
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
| | - Maria Bell
- Sanford Gynecologic Oncology, Sanford Health, 1309 West 17th St., Sioux Falls, SD 57104, USA;
| | - Mark Morgan
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Ju-Yoon Yoon
- Laboratory Medicine, Department of Pathology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St., Philadelphia, PA 19104, USA; (J.-Y.Y.); (A.B.); (L.E.S.)
| | - Marilyn A. Mitchell
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Anna Budina
- Laboratory Medicine, Department of Pathology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St., Philadelphia, PA 19104, USA; (J.-Y.Y.); (A.B.); (L.E.S.)
| | - Dalia K. Omran
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Lauren E. Schwartz
- Laboratory Medicine, Department of Pathology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St., Philadelphia, PA 19104, USA; (J.-Y.Y.); (A.B.); (L.E.S.)
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA; (N.T.); (S.S.); (M.M.); (M.A.M.); (D.K.O.); (R.D.)
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St. North, Sioux Falls, SD 57104, USA; (J.L.B.); (A.K.); (A.C.R.); (C.S.W.); (D.W.V.); (M.M.); (J.S.)
- Sanford School of Medicine, University of South Dakota, 414 East Clark St., Vermillion, SD 57069, USA
| |
Collapse
|
130
|
Scotto G, Borella F, Turinetto M, Tuninetti V, Valsecchi AA, Giannone G, Cosma S, Benedetto C, Valabrega G. Biomarkers of Central Nervous System Involvement from Epithelial Ovarian Cancer. Cells 2021; 10:3408. [PMID: 34943916 PMCID: PMC8699445 DOI: 10.3390/cells10123408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among women affected by gynaecological malignancies. Most patients show advanced disease at diagnosis (FIGO stage III-IV) and, despite the introduction of new therapeutic options, most women experience relapses. In most cases, recurrence is abdominal-pelvic; however, EOC can occasionally metastasize to distant organs, including the central nervous system. The incidence of brain metastases (BMs) from EOC is low, but it has grown over time; currently, there are no follow-up strategies available. In the last decade, a few biomarkers able to predict the risk of developing BMs from OC or as potential therapeutic targets have been investigated by several authors; to date, none have entered clinical practice. The purpose of this review is to offer a summary on the role of the most relevant predictors of central nervous system (CNS) involvement (hormone receptors; BRCA; MRD1; PD-1/PD-L1) and to highlight possible therapeutic strategies for the management of metastatic brain disease in EOC.
Collapse
Affiliation(s)
- Giulia Scotto
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Fulvio Borella
- Gynecology and Obstetrics 1, Department of Surgical Sciences, City of Health and Science, University of Turin, 10126 Turin, Italy; (F.B.); (S.C.); (C.B.)
| | - Margherita Turinetto
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Valentina Tuninetti
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Anna A. Valsecchi
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Gaia Giannone
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Stefano Cosma
- Gynecology and Obstetrics 1, Department of Surgical Sciences, City of Health and Science, University of Turin, 10126 Turin, Italy; (F.B.); (S.C.); (C.B.)
| | - Chiara Benedetto
- Gynecology and Obstetrics 1, Department of Surgical Sciences, City of Health and Science, University of Turin, 10126 Turin, Italy; (F.B.); (S.C.); (C.B.)
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, 10123 Torino, Italy; (G.S.); (M.T.); (V.T.); (A.A.V.); (G.G.)
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| |
Collapse
|
131
|
Peluso JJ, Pru JK. Progesterone Receptor Membrane Component (PGRMC)1 and PGRMC2 and Their Roles in Ovarian and Endometrial Cancer. Cancers (Basel) 2021; 13:cancers13235953. [PMID: 34885064 PMCID: PMC8656518 DOI: 10.3390/cancers13235953] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 01/02/2023] Open
Abstract
Cancers of the female reproductive tract are both lethal and highly prevalent. For example, the five-year survival rate of women diagnosed with ovarian cancer is still less than 50%, and endometrial cancer is the fourth most common cancer in women with > 65,000 new cases in the United States in 2020. Among the many genes already established as key participants in ovarian and endometrial oncogenesis, progesterone receptor membrane component (PGRMC)1 and PGRMC2 have gained recent attention given that there is now solid correlative information supporting a role for at least PGRMC1 in enhancing tumor growth and chemoresistance. The expression of PGRMC1 is significantly increased in both ovarian and endometrial cancers, similar to that reported in other cancer types. Xenograft studies using human ovarian and endometrial cancer cell lines in immunocompromised mice demonstrate that reduced expression of PGRMC1 results in tumors that grow substantially slower. While the molecular underpinnings of PGRMCs' mechanisms of action are not clearly established, it is known that PGRMCs regulate survival pathways that attenuate stress-induced cell death. The objective of this review is to provide an overview of what is known about the roles that PGRMC1 and PGRMC2 play in ovarian and endometrial cancers, particularly as related to the mechanisms through which they regulate mitosis, apoptosis, chemoresistance, and cell migration.
Collapse
Affiliation(s)
- John J. Peluso
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, CT 06030, USA
- Correspondence: ; +1-860-679-2860
| | - James K. Pru
- Department of Animal Science, Program in Reproductive Biology, University of Wyoming, Laramie, WY 82071, USA;
| |
Collapse
|
132
|
Chen R, Ishak CA, De Carvalho DD. Endogenous Retroelements and the Viral Mimicry Response in Cancer Therapy and Cellular Homeostasis. Cancer Discov 2021; 11:2707-2725. [PMID: 34649957 DOI: 10.1158/2159-8290.cd-21-0506] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/14/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022]
Abstract
Features of the cancer epigenome distinguish cancers from their respective cell of origin and establish therapeutic vulnerabilities that can be exploited through pharmacologic inhibition of DNA- or histone-modifying enzymes. Epigenetic therapies converge with cancer immunotherapies through "viral mimicry," a cellular state of active antiviral response triggered by endogenous nucleic acids often derived from aberrantly transcribed endogenous retrotransposons. This review describes the initial characterization and expansion of viral mimicry-inducing approaches as well as features that "prime" cancers for viral mimicry induction. Increased understanding of viral mimicry in therapeutic contexts suggests potential physiologic roles in cellular homeostasis. SIGNIFICANCE: Recent literature establishes elevated cytosolic double strand RNA (dsRNA) levels as a cancer-specific therapeutic vulnerability that can be elevated by viral mimicry-inducing therapies beyond tolerable thresholds to induce antiviral signaling and increase dependence on dsRNA stress responses mediated by ADAR1. Improved understanding of viral mimicry signaling and tolerance mechanisms reveals synergistic treatment combinations with epigenetic therapies that include inhibition of BCL2, ADAR1, and immune checkpoint blockade. Further characterization of viral mimicry tolerance may identify contexts that maximize efficacy of conventional cancer therapies.
Collapse
Affiliation(s)
- Raymond Chen
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Charles A Ishak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Daniel D De Carvalho
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
133
|
Zeng H, Chen L, Zhang M, Luo Y, Ma X. Integration of histopathological images and multi-dimensional omics analyses predicts molecular features and prognosis in high-grade serous ovarian cancer. Gynecol Oncol 2021; 163:171-180. [PMID: 34275655 DOI: 10.1016/j.ygyno.2021.07.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study used histopathological image features to predict molecular features, and combined with multi-dimensional omics data to predict overall survival (OS) in high-grade serous ovarian cancer (HGSOC). METHODS Patients from The Cancer Genome Atlas (TCGA) were distributed into training set (n = 115) and test set (n = 114). In addition, we collected tissue microarrays of 92 patients as an external validation set. Quantitative features were extracted from histopathological images using CellProfiler, and utilized to establish prediction models by machine learning methods in training set. The prediction performance was assessed in test set and validation set. RESULTS The prediction models were able to identify BRCA1 mutation (AUC = 0.952), BRCA2 mutation (AUC = 0.912), microsatellite instability-high (AUC = 0.919), microsatellite stable (AUC = 0.924), and molecular subtypes: proliferative (AUC = 0.961), differentiated (AUC = 0.952), immunoreactive (AUC = 0.941), mesenchymal (AUC = 0.918) in test set. The prognostic model based on histopathological image features could predict OS in test set (5-year AUC = 0.825) and validation set (5-year AUC = 0.703). We next explored the integrative prognostic models of image features, genomics, transcriptomics and proteomics. In test set, the models combining two omics had higher prediction accuracy, such as image features and genomics (5-year AUC = 0.834). The multi-omics model including all features showed the best prediction performance (5-year AUC = 0.911). According to risk score of multi-omics model, the high-risk and low-risk groups had significant survival differences (HR = 18.23, p < 0.001). CONCLUSIONS These results indicated the potential ability of histopathological image features to predict above molecular features and survival risk of HGSOC patients. The integration of image features and multi-omics data may improve prognosis prediction in HGSOC patients.
Collapse
Affiliation(s)
- Hao Zeng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, China
| | - Linyan Chen
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, China
| | - Mingxuan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuling Luo
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
134
|
Cordova R, Kiekens K, Burrell S, Drake W, Kmeid Z, Rice P, Rocha A, Diaz S, Yamada S, Yozwiak M, Nelson OL, Rodriguez GC, Heusinkveld J, Shih IM, Alberts DS, Barton JK. Sub-millimeter endoscope demonstrates feasibility of in vivo reflectance imaging, fluorescence imaging, and cell collection in the fallopian tubes. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-200404R. [PMID: 34216135 PMCID: PMC8253554 DOI: 10.1117/1.jbo.26.7.076001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/15/2021] [Indexed: 06/13/2023]
Abstract
SIGNIFICANCE Most cases of high-grade serous ovarian carcinoma originate as serous tubal intraepithelial carcinoma (STIC) lesions in the fallopian tube epithelium (FTE), enabling early endoscopic detection. AIM The cell-acquiring fallopian endoscope (CAFE) was built to meet requirements for locating potentially pathological tissue indicated by an alteration in autofluorescence or presence of a targeted fluorophore. A channel was included for directed scrape biopsy of cells from regions of interest. APPROACH Imaging resolution and fluorescence sensitivity were measured using a standard resolution target and fluorescence standards, respectively. A prototype was tested in ex vivo tissue, and collected cells were counted and processed. RESULTS Measured imaging resolution was 88 μm at a 5-mm distance, and full field of view was ∼45 deg in air. Reflectance and fluorescence images in ex vivo porcine reproductive tracts were captured, and fit through human tracts was verified. Hemocytometry counts showed that on the order of 105 cells per scrape biopsy could be collected from ex vivo porcine tissue. CONCLUSIONS All requirements for viewing STIC in the FTE were met, and collected cell counts exceeded input requirements for relevant analyses. Our benchtop findings suggest the potential utility of the CAFE device for in vivo imaging and cell collection in future clinical trials.
Collapse
Affiliation(s)
- Ricky Cordova
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Kelli Kiekens
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Susan Burrell
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - William Drake
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Zaynah Kmeid
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Photini Rice
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Andrew Rocha
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Sebastian Diaz
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Shigehiro Yamada
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland, United States
| | - Michael Yozwiak
- University of Arizona, Department of Medicine, Tucson, Arizona, United States
| | - Omar L. Nelson
- NorthShore University HealthSystem, Evanston, Illinois, United States
- University of Chicago, Pritzker School of Medicine, Chicago, Illinois, United States
| | - Gustavo C. Rodriguez
- NorthShore University HealthSystem, Evanston, Illinois, United States
- University of Chicago, Pritzker School of Medicine, Chicago, Illinois, United States
| | - John Heusinkveld
- Banner–University Medical Center, Tucson, Arizona, United States
| | - Ie-Ming Shih
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland, United States
| | - David S. Alberts
- University of Arizona, Department of Medicine, Tucson, Arizona, United States
| | - Jennifer K. Barton
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| |
Collapse
|
135
|
Luo X, Xu J, Yu J, Yi P. Shaping Immune Responses in the Tumor Microenvironment of Ovarian Cancer. Front Immunol 2021; 12:692360. [PMID: 34248988 PMCID: PMC8261131 DOI: 10.3389/fimmu.2021.692360] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Reciprocal signaling between immune cells and ovarian cancer cells in the tumor microenvironment can alter immune responses and regulate disease progression. These signaling events are regulated by multiple factors, including genetic and epigenetic alterations in both the ovarian cancer cells and immune cells, as well as cytokine pathways. Multiple immune cell types are recruited to the ovarian cancer tumor microenvironment, and new insights about the complexity of their interactions have emerged in recent years. The growing understanding of immune cell function in the ovarian cancer tumor microenvironment has important implications for biomarker discovery and therapeutic development. This review aims to describe the factors that shape the phenotypes of immune cells in the tumor microenvironment of ovarian cancer and how these changes impact disease progression and therapy.
Collapse
Affiliation(s)
- Xin Luo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States.,Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
136
|
Mori S, Gotoh O, Kiyotani K, Low SK. Genomic alterations in gynecological malignancies: histotype-associated driver mutations, molecular subtyping schemes, and tumorigenic mechanisms. J Hum Genet 2021; 66:853-868. [PMID: 34092788 DOI: 10.1038/s10038-021-00940-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/14/2021] [Accepted: 05/25/2021] [Indexed: 02/08/2023]
Abstract
There are numerous histological subtypes (histotypes) of gynecological malignancies, with each histotype considered to largely reflect a feature of the "cell of origin," and to be tightly linked with the clinical behavior and biological phenotype of the tumor. The recent advances in massive parallel sequencing technologies have provided a more complete picture of the range of the genomic alterations that can persist within individual tumors, and have highlighted the types and frequencies of driver-gene mutations and molecular subtypes often associated with these histotypes. Several large-scale genomic cohorts, including the Cancer Genome Atlas (TCGA), have been used to characterize the genomic features of a range of gynecological malignancies, including high-grade serous ovarian carcinoma, uterine corpus endometrial carcinoma, uterine cervical carcinoma, and uterine carcinosarcoma. These datasets have also been pivotal in identifying clinically relevant molecular targets and biomarkers, and in the construction of molecular subtyping schemes. In addition, the recent widespread use of clinical sequencing for the more ubiquitous types of gynecological cancer has manifested in a series of large genomic datasets that have allowed the characterization of the genomes, driver mutations, and histotypes of even rare cancer types, with sufficient statistical power. Here, we review the field of gynecological cancer, and seek to describe the genomic features by histotype. We also will demonstrate how these are linked with clinicopathological attributes and highlight the potential tumorigenic mechanisms.
Collapse
Affiliation(s)
- Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Osamu Gotoh
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Siew Kee Low
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
137
|
De Leo A, Santini D, Ceccarelli C, Santandrea G, Palicelli A, Acquaviva G, Chiarucci F, Rosini F, Ravegnini G, Pession A, Turchetti D, Zamagni C, Perrone AM, De Iaco P, Tallini G, de Biase D. What Is New on Ovarian Carcinoma: Integrated Morphologic and Molecular Analysis Following the New 2020 World Health Organization Classification of Female Genital Tumors. Diagnostics (Basel) 2021; 11:697. [PMID: 33919741 PMCID: PMC8070731 DOI: 10.3390/diagnostics11040697] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/05/2021] [Accepted: 04/10/2021] [Indexed: 02/05/2023] Open
Abstract
Ovarian carcinomas represent a heterogeneous group of neoplasms consisting of separate entities with distinct risk factors, precursor lesions, pathogenesis, patterns of spread, molecular profiles, clinical course, response to chemotherapy, and outcomes. The histologic subtype and the related molecular features are essential for individualized clinical decision-making. The fifth edition of the World Health Organization classification of tumors of the female genital tract divides ovarian carcinomas into at least five main and distinct types of ovarian carcinomas: high-grade serous carcinoma, low-grade serous carcinoma, endometrioid carcinoma, clear cell carcinoma, and mucinous carcinoma. Molecular pathology has improved the knowledge of genomic landscape of ovarian carcinomas identifying peculiar alterations for every histologic subtype. It is well-known that high-grade and low-grade serous carcinomas are separate entities with entirely different morphologic and molecular characteristics. TP53 and BRCA mutations are typical of high-grade serous carcinoma, whereas BRAF and KRAS mutations frequently occur in low-grade serous carcinoma. Endometrioid and clear cell carcinomas are frequently associated with endometriosis. Endometrioid tumors are characterized by β-catenin alterations, microsatellite instability, and PTEN and POLE mutations, while ARID1A mutations occur in both endometrioid and clear cell carcinomas. Mucinous carcinomas are uncommon tumors associated with copy-number loss of CDKN2A and KRAS alterations and metastasis from other sites should always be considered in the differential diagnosis.
Collapse
Affiliation(s)
- Antonio De Leo
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum—University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (C.C.); (G.A.); (F.C.); (G.T.)
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
| | - Donatella Santini
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Pathology Unit, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Claudio Ceccarelli
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum—University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (C.C.); (G.A.); (F.C.); (G.T.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
| | - Giacomo Santandrea
- Pathology Unit, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (A.P.)
| | - Andrea Palicelli
- Pathology Unit, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (A.P.)
| | - Giorgia Acquaviva
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum—University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (C.C.); (G.A.); (F.C.); (G.T.)
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
| | - Federico Chiarucci
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum—University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (C.C.); (G.A.); (F.C.); (G.T.)
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
| | - Francesca Rosini
- Pathology Unit, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Gloria Ravegnini
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Annalisa Pession
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Daniela Turchetti
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Claudio Zamagni
- IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy;
| | - Anna Myriam Perrone
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Massarenti 13, 40138 Bologna, Italy
| | - Pierandrea De Iaco
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Via Massarenti 13, 40138 Bologna, Italy
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum—University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (C.C.); (G.A.); (F.C.); (G.T.)
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
| | - Dario de Biase
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero—Universitaria di Bologna/Azienda USL di Bologna, 40138 Bologna, Italy; (A.P.); (D.d.B.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy; (D.S.); (G.R.); (D.T.); (A.M.P.); (P.D.I.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
138
|
Yachida N, Yoshihara K, Yamaguchi M, Suda K, Tamura R, Enomoto T. How Does Endometriosis Lead to Ovarian Cancer? The Molecular Mechanism of Endometriosis-Associated Ovarian Cancer Development. Cancers (Basel) 2021; 13:1439. [PMID: 33809880 PMCID: PMC8004227 DOI: 10.3390/cancers13061439] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Numerous epidemiological and histopathological studies support the notion that clear cell and endometrioid carcinomas derive from ovarian endometriosis. Accordingly, these histologic types are referred to as "endometriosis-associated ovarian cancer" (EAOC). Although the uterine endometrium is also considered an origin of endometriosis, the molecular mechanism involved in transformation of the uterine endometrium to EAOC via ovarian endometriosis has not yet been clarified. Recent studies based on high-throughput sequencing technology have revealed that cancer-associated gene mutations frequently identified in EAOC may exist in the normal uterine endometrial epithelium and ovarian endometriotic epithelium. The continuum of genomic alterations from the uterine endometrium to endometriosis and EAOC has been described, though the significance of cancer-associated gene mutations in the uterine endometrium or endometriosis remains unclear. In this review, we summarize current knowledge regarding the molecular characteristics of the uterine endometrium, endometriosis, and EAOC and discuss the molecular mechanism of cancer development from the normal endometrium through endometriosis in an effort to prevent EAOC.
Collapse
Affiliation(s)
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (N.Y.); (M.Y.); (K.S.); (R.T.); (T.E.)
| | | | | | | | | |
Collapse
|
139
|
Bacterial, Archaea, and Viral Transcripts (BAVT) Expression in Gynecological Cancers and Correlation with Regulatory Regions of the Genome. Cancers (Basel) 2021; 13:cancers13051109. [PMID: 33807612 PMCID: PMC7961894 DOI: 10.3390/cancers13051109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/31/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Microorganisms are found in all human tissues. Some of them are responsible for cancer formation. In our study we found gene expression from bacteria, archaea, and viruses in the upper female genital tract and this expression was associated with ovarian and endometrial cancer. We also found that the expression from these organisms may be involved in regulatory mechanisms of infection and cancer formation. Some of the processes associated with these organisms may affect cancer heterogeneity and be potential targets for cancer therapy. Abstract Bacteria, archaea, and viruses are associated with numerous human cancers. To date, microbiome variations in transcription have not been evaluated relative to upper female genital tract cancer risk. Our aim was to assess differences in bacterial, archaea, and viral transcript (BAVT) expression between different gynecological cancers and normal fallopian tubes. In this case-control study we performed RNA sequencing on 12 normal tubes, 112 serous ovarian cancers (HGSC) and 62 endometrioid endometrial cancers (EEC). We used the centrifuge algorithm to classify resultant transcripts into four indexes: bacterial, archaea, viral, and human genomes. We then compared BAVT expression from normal samples, HGSC and EEC. T-test was used for univariate comparisons (correcting for multiple comparison) and lasso for multivariate modelling. For validation we performed DNA sequencing of normal tubes in comparison to HGSC and EEC BAVTs in the TCGA database. Pathway analyses were carried out to evaluate the function of significant BAVTs. Our results show that BAVT expression levels vary between different gynecological cancers. Finally, we mapped some of these BAVTs to the human genome. Numerous map locations were close to regulatory genes and long non-coding RNAs based on the pathway enrichment analysis. BAVTs may affect gynecological cancer risk and may be part of potential targets for cancer therapy.
Collapse
|
140
|
Ding DC, Chang YH, Wu KC, Harnod T. The organoid: A research model for ovarian cancer. Tzu Chi Med J 2021; 34:255-260. [PMID: 35912056 PMCID: PMC9333109 DOI: 10.4103/tcmj.tcmj_63_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a heterogeneous disease with a variety of distinct clinical and molecular characteristics. The currently available and common research models for EOC include tumor cell lines and patient-derived xenografts. However, these models have certain shortcomings: establishing a cell line is time-consuming, loss of genetic traits after long-term culture is a possibility, and investment is required in terms of animal care facilities. Therefore, better research models are required. Organoid technology was originally developed from colorectal cancer. Tumor organoid is a three-dimensional culture system and can help accurately recapture the tumor phenotype from the original tumor. Tumor organoid systems can overcome the above-mentioned shortcomings of the currently available research models. The organoid model can be used for culturing ovarian cancer subtypes, screening drugs, assessing genomes, and establishing biobanks. However, the currently available organoid models can only culture one type of cells, epithelial cells. Therefore, an organoid-on-a-chip device can be developed in the future to provide a microenvironment for cell–cell, cell–matrix, and cell–media interactions. Thus, organoid models can be used in ovarian cancer research and can generate a simulated in vivo system, enabling studies on the heterogeneity of ovarian cancer.
Collapse
|
141
|
Sánchez-Borrego R, Sánchez-Prieto M. What are the mechanisms of action of the different contraceptive methods to reduce the risk of ovarian cancer? EUR J CONTRACEP REPR 2020; 26:79-84. [PMID: 33245019 DOI: 10.1080/13625187.2020.1849617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Ovarian cancer (OvCa) is the deadliest gynaecologic malignancy. Knowing that OvCa, as a disease, has different origins has allowed us to relate them to the mechanisms of action of different contraceptive methods with the aim of evaluating the possibility of their use in reducing risk. STUDY DESIGN This commentary review article will instead focus on the recent findings on the role of contraceptive methods in preventing of OvCa. RESULTS Combined hormonal contraceptive (CHC) use is an effective method of chemoprevention for OvCa in the general population and in women with genetic disorders. Salpingectomy, better than tubal ligation, should be offered for ovarian/tubal/peritoneal cancer prevention. Progestogen-only methods can decrease the risk of OvCa via reduced menstrual bleeding and by changes in the hormonal environment that surrounds the ovary. IUDs of any type, through different mechanisms, decrease the risk of OvCa. Barrier methods prevent the passage of germs into the tubes and ovaries and the inflammatory state they produce. CONCLUSIONS Most contraceptive methods have a mechanism of action that may favour a reduction in the risk of OvCa. The theories of incessant ovulation, retrograde menstruation, and that the fallopian tubes are the site of origin of a proportion of high-grade serous OvCa, have led to the recommendation that anovulatory methods, those that decrease menstrual bleeding, and those that blocked tubes, or even better, 'opportunistic salpingectomy' are a current approach to prevent OvCa in the population general and, above all, in the population at risk.
Collapse
Affiliation(s)
| | - Manuel Sánchez-Prieto
- Department of Obstetrics and Gynecology, Institut Universitari Dexeus, Barcelona, Spain
| |
Collapse
|