1
|
Rushton T, Krause HB, Samec T, Elliott A, Karnezis AN, Toboni MD, Thaker PH, Braxton DR, Oberley M, Gershenson DM, Armstrong DK. Characterizing the genomic landscape through the lens of FOLR1 status in low and high grade serous ovarian carcinoma. Gynecol Oncol 2024; 191:80-85. [PMID: 39366033 DOI: 10.1016/j.ygyno.2024.09.021] [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: 07/11/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVE Targeted therapy in folate receptor alpha (FOLR1)-positive high grade serous ovarian carcinoma (HGSOC) is now a mainstay for platinum-resistant disease. However, the rate of FOLR1-positivity in low grade serous ovarian carcinoma (LGSOC) is not well documented. Less common than HGSOC, LGSOC tends to respond poorly to traditional platinum-based chemotherapeutic regimens, particularly in recurrence. Thus, there is an urgent need to identify molecular targets that may assist in identifying more efficacious treatments for LGSOC. In this work, we assessed the genomic and transcriptomic landscapes in FOLR1-positive/negative LGSOC compared to its high-grade counterpart. METHODS Using a large precision oncology database, next-generation sequencing and immunohistochemistry was performed on a cohort of 281 LGSOC and 5086 HGSOC. Associated MAPK activation was calculated based on NGS results and patient survival analysis was completed stratified by molecular alteration. RESULTS Compared with LGSOC (24.6 %), HGSOC tumors have significantly higher prevalence of FOLR1+ status (43.5 %) and significantly higher PD-L1+ status. Conversely, LGSOC had higher prevalence of KRAS and NRAS mutations, with a near exclusivity for BRAF mutation compared to HGSOC. FOLR1- LGSOC and HGSOC had similar prevalences of T cell-inflamed tumors, though FOLR1+ LGSOC had a significantly lower prevalence of T-Cell inflamed tumors than FOLR1+ HGSOC. MAPK activation, quantified via MAPK activation score (MPAS), was significantly higher in low-grade tumors compared to HGSOC, yet no difference between FOLR1+ vs FOLR1- LGSOC was observed. CONCLUSIONS Though less than in high-grade disease, a notable portion of low-grade tumors were FOLR1+, suggesting FOLR1 expression in LGSOC could be a viable target for this rare histology, particularly in the recurrent setting.
Collapse
Affiliation(s)
- Tullia Rushton
- Johns Hopkins University, Baltimore, MD, United States of America.
| | | | | | | | - Anthony N Karnezis
- University of California-Davis Health, Sacramento, CA, United States of America
| | - Michael D Toboni
- University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Premal H Thaker
- Washington University in St. Lous, St. Louis, MO, United States of America
| | - David R Braxton
- Hoag Family Cancer Institute and Hoag Health, Newport Beach, CA, United States of America
| | | | - David M Gershenson
- University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | | |
Collapse
|
2
|
Li Y, Gan X, Li F, Hu L. The Putative Effects of Neoadjuvant Chemotherapy on the Immune System of Advanced Epithelial Ovarian Carcinoma. Immunol Invest 2024; 53:91-114. [PMID: 37987679 DOI: 10.1080/08820139.2023.2284885] [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] [Indexed: 11/22/2023]
Abstract
The epithelial ovarian carcinoma (EOC) is one of leading causes of cancer-related mortality in females. For some patients, complete resection cannot be achieved, thus neoadjuvant chemotherapy (NACT) following interval debulking surgery (IDS) could be an alternative choice. In general-held belief, cytotoxic chemotherapy is assumed to be immunosuppressive, because of its toxicity to dividing cells in the bone marrow and peripheral lymphoid tissues. However, increasing evidence highlighted that the anticancer activity of chemotherapy may also be related to its ability to act as an immune modulator. NACT not only changed the morphology of cancer cells, but also changed the transcriptomic and genomic profile of EOC, induced proliferation of cancer stem-like cells, gene mutation, and tumor-related adaptive immune response. This review will provide a comprehensive overview of recent studies evaluating the impact of NACT on cancer cells and immune system of advanced EOC and their relationship to clinical outcome. This information could help us understand the change of immune system during NACT, which might provide new strategies in future investigation of immuno-therapy for maintenance treatment of EOC.
Collapse
Affiliation(s)
- Yunyun Li
- Department of Gynecology and Obstetrics, The Yongchuan Hospital of Chongqing Medical University, Yongchuan District, Chongqing, PR China
- Department of Gynecology, Second Affiliated Hospital of Chongqing Medical University, Nanan District, Chongqing, PR China
| | - Xiaoling Gan
- Department of Gynecology, Second Affiliated Hospital of Chongqing Medical University, Nanan District, Chongqing, PR China
| | - Fei Li
- Department of Gynecology and Obstetrics, The Yongchuan Hospital of Chongqing Medical University, Yongchuan District, Chongqing, PR China
| | - Lina Hu
- Department of Gynecology, Second Affiliated Hospital of Chongqing Medical University, Nanan District, Chongqing, PR China
| |
Collapse
|
3
|
McKenzie ND, Ahmad S, Awada A, Kuhn TM, Recio FO, Holloway RW. Prognostic features of the tumor microenvironment in high-grade serous ovarian cancer and dietary immunomodulation. Life Sci 2023; 333:122178. [PMID: 37839778 DOI: 10.1016/j.lfs.2023.122178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is a particularly lethal malignancy that is prognostically influenced by the immune profile of the tumor microenvironment (TME). TME immune profiles have been sub-categorized according to features associated with both survival outcomes as well as response to systemic therapies. Five suggested immune phenotypes have been described and correlated with overall survival outcomes. Phenotypes associated with shorter overall survival rates appear to have prominent immunosuppressive features within their TME. The opportunity to triage patients according to their prognostic TME profile might allow selection of individual patients with poor prognostic features who could most benefit from innovative immunomodulatory treatment strategies. Two potential strategies to indirectly manipulate the TME (and oncologic outcomes) are alteration of the gut microbiome composition and alteration of TME metabolism through dietary interventions. Experimental dietary modifications in humans designed for influencing cancer outcomes are only beginning to be studied in a prospective fashion. Herein we summarize prognostic TME features in HGSOC and potential opportunities for immunomodulation via dietary and gut microbial interventions.
Collapse
Affiliation(s)
- Nathalie D McKenzie
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Ahmad Awada
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Theresa M Kuhn
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Fernando O Recio
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Robert W Holloway
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| |
Collapse
|
4
|
Fei H, Han X, Wang Y, Li S. Novel immune-related gene signature for risk stratification and prognosis prediction in ovarian cancer. J Ovarian Res 2023; 16:205. [PMID: 37858138 PMCID: PMC10585734 DOI: 10.1186/s13048-023-01289-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 09/28/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The immune system played a multifaceted role in ovarian cancer (OC) and was a significant mediator of ovarian carcinogenesis. Various immune cells and immune gene products played an integrated role in ovarian cancer (OC) progression, proved the significance of the immune microenvironment in prognosis. Therefore, we aimed to establish and validate an immune gene prognostic signature for OC patients' prognosis prediction. METHODS Differently expressed Immune-related genes (DEIRGs) were identified in 428 OC and 77 normal ovary tissue specimens from 9 independent GEO datasets. The Cancer Genome Atlas (TCGA) cohort was used as a training cohort, Univariate Cox analysis was used to identify prognostic DEIRGs in TCGA cohort. Then, an immune gene-based risk model for prognosis prediction was constructed using the LASSO regression analysis, and validated the accuracy and stability of the model in 374 and 93 OC patients in TCGA training cohort and International Cancer Genome Consortium (ICGC) validation cohort respectively. Finally, the correlation among risk score model, clinicopathological parameters, and immune cell infiltration were analyzed. RESULTS Five DEIRGs were identified to establish the immune gene signature and divided OC patients into the low- and high-risk groups. In TCGA and ICGC datasets, patients in the low-risk group showed a substantially higher survival rate than high-risk group. Receiver operating characteristic (ROC) curves, t-distributed stochastic neighbor embedding (t-SNE) analysis and principal component analysis (PCA) showed the good performance of the risk model. Clinicopathological correlation analysis proved the risk score model could serve as an independent prognostic factor in 2 independent datasets. CONCLUSIONS The prognostic model based on immune-related genes can function as a superior prognostic indicator for OC patients, which could provide evidence for individualized treatment and clinical decision making.
Collapse
Affiliation(s)
- Hongjun Fei
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Municipal Key Clinical Specialty, Shanghai Jiao Tong University School of Medicine, No.910, Hengshan Road, Shanghai, 200030, China.
| | - Xu Han
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Municipal Key Clinical Specialty, Shanghai Jiao Tong University School of Medicine, No.910, Hengshan Road, Shanghai, 200030, China
| | - Yanlin Wang
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Municipal Key Clinical Specialty, Shanghai Jiao Tong University School of Medicine, No.910, Hengshan Road, Shanghai, 200030, China
| | - Shuyuan Li
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Municipal Key Clinical Specialty, Shanghai Jiao Tong University School of Medicine, No.910, Hengshan Road, Shanghai, 200030, China.
| |
Collapse
|
5
|
Said SA, de Hullu JA, van der Aa MA, Walraven JEW, Bekkers RLM, Slangen BFM, Pickkers P, van Altena AM. Impact of Sepsis on the Oncologic Outcomes of Advanced Epithelial Ovarian Cancer Patients: A Multicenter Observational Study. Cancers (Basel) 2023; 15:4642. [PMID: 37760610 PMCID: PMC10526225 DOI: 10.3390/cancers15184642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE The sepsis-induced inflammatory response may potentially affect malignant cells. Recently, a case of spontaneous regression of a histologically confirmed International Federation of Gynecology and Obstetrics (FIGO) stage IIIC epithelial ovarian cancer (EOC) following sepsis was reported. The aim of our study was to assess the impact of sepsis on the oncologic outcomes of advanced-stage EOC patients. METHODS Gynecologic oncologic patients admitted to the Intensive Care Unit of three oncologic centers between 2006 and 2019 were identified and patients who experienced sepsis following advanced-stage EOC diagnosis were selected. Survival outcomes were compared with advanced-stage EOC patients from the Netherlands Cancer Registry (NCR). To correct for case-mix differences, propensity score matching using 1:3 nearest neighbor matching was conducted after which survival analyses were repeated. RESULTS A total of 18 of 215 patients with advanced-stage EOC experienced sepsis. Sepsis patients had similar distributions of patient, tumor, and treatment characteristics to 3988 patients from the NCR cohort. A total of 3 of 18 patients died from the complications of sepsis. While the remaining patients initially responded to treatment, 14/15 patients relapsed. The median (IQR) overall survival was 31 (24-44) and 35 (20-60) months for the sepsis and unmatched NCR cohort (p = 0.56), respectively. The median (IQR) progression-free survival was 16 (11-21) and 16 (11-27) months (p = 0.90), respectively. Survival outcomes did not differ following propensity matching (overall survival of 31 (24-44) vs. 36 (20-56) months, p = 0.40; progression-free survival of 16 (11-21) and 16 (12-21) months, p = 0.72). CONCLUSION In this observational study, the occurrence of sepsis did not affect the oncologic and survival outcomes of advanced-stage EOC patients.
Collapse
Affiliation(s)
- Sherin A. Said
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Joanne A. de Hullu
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| | - Maaike A. van der Aa
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Janneke E. W. Walraven
- Department of Medical Oncology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands
| | - Ruud L. M. Bekkers
- Department of Obstetrics and Gynecology, Catharina Hospital, 5623 EJ Eindhoven, The Netherlands
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
| | - Brigitte F. M. Slangen
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands;
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| |
Collapse
|
6
|
Cai D, Liu T, Fang J, Liu Y. Molecular cluster mining of high-grade serous ovarian cancer via multi-omics data analysis aids precise medicine. J Cancer Res Clin Oncol 2023; 149:9151-9165. [PMID: 37178426 DOI: 10.1007/s00432-023-04831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
PURPOSE HGSOC is a kind of gynecological cancer with high mortality and strong heterogeneity. The study used multi-omics and multiple algorithms to identify novel molecular subtypes, which can help patients obtain more personalized treatments. METHODS Firstly, the consensus clustering result was obtained using a consensus ensemble of ten classical clustering algorithms, based on mRNA, lncRNA, DNA methylation, and mutation data. The difference in signaling pathways was evaluated using the single-sample gene set enrichment analysis (ssGSEA). Meanwhile, the relationship between genetic alteration, response to immunotherapy, drug sensitivity, prognosis, and subtypes was further analyzed. Finally, the reliability of the new subtype was verified in three external datasets. RESULTS Three molecular subtypes were identified. Immune desert subtype (CS1) had little enrichment in the immune microenvironment and metabolic pathways. Immune/non-stromal subtype (CS2) was enriched in the immune microenvironment and metabolism of polyamines. Immune/stromal subtype (CS3) not only enriched anti-tumor immune microenvironment characteristics but also enriched pro-tumor stroma characteristics, glycosaminoglycan metabolism, and sphingolipid metabolism. The CS2 had the best overall survival and the highest response rate to immunotherapy. The CS3 had the worst prognosis and the lowest response rate to immunotherapy but was more sensitive to PARP and VEGFR molecular-targeted therapy. The similar differences among three subtypes were successfully validated in three external cohorts. CONCLUSION We used ten clustering algorithms to comprehensively analyze four types of omics data, identified three biologically significant subtypes of HGSOC patients, and provided personalized treatment recommendations for each subtype. Our findings provided novel views into the HGSOC subtypes and could provide potential clinical treatment strategies.
Collapse
Affiliation(s)
- Daren Cai
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Tiantian Liu
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Jingya Fang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China.
| | - Yingbo Liu
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
7
|
Wang YM, Cai W, Xue QM, Zhang JY, Zhou L, Xiong SY, Deng H. Prognostic role of different PD-L1 expression patterns and tumor-infiltrating lymphocytes in high-grade serous ovarian cancer: a systematic review and meta-analysis. Front Immunol 2023; 14:1234894. [PMID: 37654479 PMCID: PMC10465691 DOI: 10.3389/fimmu.2023.1234894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Background The prognostic value of programmed cell death ligand 1 (PD-L1) expression and tumor-infiltrating lymphocytes (TILs) in high-grade serous ovarian cancer (HGSOC) remains a controversial topic in the research field. To comprehensively assess the importance of PD-L1 and TILs in this particular subtype of ovarian cancer, we performed a meta-analysis. Methods We conducted a comprehensive search of PubMed, Embase, Scopus, Web of Science, and Cochrane Library databases up to December 25, 2022. The association between PD-L1, TILs, and survival outcomes was evaluated using the combined hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs). Results This meta-analysis comprised 11 trials involving a total of 1746 cases. The results revealed no significant association between PD-L1 expression in tumor cells (TCs) and overall survival (OS, HR = 0.76, 95% CI: 0.52-1.09, p = 0.136) or progression-free survival (PFS, HR = 0.71, 95% CI: 0.4 -1.24, p = 0.230). Nevertheless, a correlation was observed between PD-L1 expression in immune cells (ICs) and OS (HR = 0.73, 95% CI: 0.55-0.97, p = 0.031). Furthermore, the presence of CD8+ and PD-1+ TILs was found to significantly enhance OS (HR = 0.70, 95% CI = 0.55-0.87, p = 0.002; HR = 0.57, 95% CI = 0.40-0.80, p = 0.001, respectively) and PFS (HR = 0.62, 95% CI = 0.41-0.92, p = 0.019; HR = 0.52, 95% CI = 0.35-0.78, p = 0.002, respectively), whereas the presence of CD3+ and CD4+ TILs was positively associated with OS (HR = 0.50, 95% CI = 0.29-0.87, p = 0.014; HR = 0.55, 95% CI = 0.34-0.91, p = 0.020, respectively). Conclusion This study indicates a positive correlation between ICs-derived PD-L1 and survival, while no significant correlation was observed between TCs-derived PD-L1 and prognosis. These results highlight the importance of studying PD-L1 expression in ICs as a prognostic predictor. In addition, the presence of TILs was found to significantly improve patient survival, suggesting that TILs may be a valuable prognostic biomarker. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022366411.
Collapse
Affiliation(s)
- Ye-Min Wang
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Cai
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qing-Ming Xue
- Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jin-Yao Zhang
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lv Zhou
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Su-Yi Xiong
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huan Deng
- Department of Pathology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Wang C, Yu C, Chang H, Song J, Zhang S, Zhao J, Wang J, Wang T, Qi Q, Shan C. Glucose-6-phosphate dehydrogenase: a therapeutic target for ovarian cancer. Expert Opin Ther Targets 2023; 27:733-743. [PMID: 37571851 DOI: 10.1080/14728222.2023.2247558] [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: 02/13/2023] [Revised: 07/04/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023]
Abstract
INTRODUCTION Ovarian cancer (OC) is a gynecological tumor disease, which is usually diagnosed at an advanced stage and has a poor prognosis. It has been established that the glucose metabolism rate of cancer cells is significantly higher than that of normal cells, and the pentose phosphate pathway (PPP) is an important branch pathway for glucose metabolism. Glucose-6-phosphate dehydrogenase (G6PD) is the key rate-limiting enzyme in the PPP, which plays an important role in the initiation and development of cancer (such as OC), and has been considered as a promisinganti-cancer target. AREAS COVERED In this review, based on the structure and biological function of G6PD, recent research on the roles of G6PD in the progression, metastasis, and chemoresistance of OC are summarized and accompanied by proposed molecular mechanisms, which may provide a systematic understanding of targeting G6PD for the treatment of patients with OC. EXPERT OPINION Accumulating evidence demonstrates that G6PD is a promising target of cancer. The development of G6PD inhibitors for cancer treatment merits broad application prospects.
Collapse
Affiliation(s)
- Chenxi Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Chenxi Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Hongkai Chang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Jiaqi Song
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Shuai Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianguo Zhao
- Tianjin Key Laboratory of human development and reproductive regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Jiyan Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Tao Wang
- Tianjin Key Laboratory of human development and reproductive regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Qi Qi
- MOE Key Laboratory of Tumor Molecular Biology, Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Tu M, Xu J. Advances in immunotherapy for gynecological malignancies. Crit Rev Oncol Hematol 2023:104063. [PMID: 37385307 DOI: 10.1016/j.critrevonc.2023.104063] [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: 02/11/2023] [Revised: 05/21/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023] Open
Abstract
To date, surgery, chemotherapy and radiotherapy are mainly used to treat or remove gynecological malignancies. However, these approaches have their limitations when facing complicated female diseases such as advanced cervical and endometrial cancer (EC), chemotherapy-resistant gestational trophoblastic neoplasia and platinum-resistant ovarian cancer. Instead, immunotherapy, as an alternative, could significantly improve prognosis of those patients receiving traditional treatments, with better antitumor activities and possibly less cellular toxicities. Its' development is still not fast enough to meet the current clinical needs. More preclinical studies and larger-scale clinical trials are required. This review aims to introduce the landscape and up-to-date status of immunotherapy against gynecological malignancies, with a discussion of the challenges and future direction.
Collapse
Affiliation(s)
- Mengyan Tu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
10
|
Berckmans Y, Hoffert Y, Vankerckhoven A, Dreesen E, Coosemans A. Drug Repurposing for Targeting Myeloid-Derived Suppressor-Cell-Generated Immunosuppression in Ovarian Cancer: A Literature Review of Potential Candidates. Pharmaceutics 2023; 15:1792. [PMID: 37513979 PMCID: PMC10385967 DOI: 10.3390/pharmaceutics15071792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The lethality of patients with ovarian cancer (OC) remains high. Current treatment strategies often do not lead to the desired outcome due to the development of therapy resistance, resulting in high relapse rates. Additionally, clinical trials testing immunotherapy against OC have failed to reach significant results to date. The OC tumor microenvironment and specifically myeloid-derived suppressor cells (MDSC) are known to generate immunosuppression and inhibit the anti-tumor immune response following immunotherapy treatment. Our review aims to characterize potential candidate treatments to target MDSC in OC through drug-repurposing. A literature search identified repurposable compounds with evidence of their suppressing the effect of MDSC. A total of seventeen compounds were withheld, of which four were considered the most promising. Lurbinectedin, metformin, celecoxib, and 5-azacytidine have reported preclinical effects on MDSC and clinical evidence in OC. They have all been approved for a different indication, characterizing them as the most promising candidates for repurposing to treat patients with OC.
Collapse
Affiliation(s)
- Yani Berckmans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Yannick Hoffert
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Ann Vankerckhoven
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Erwin Dreesen
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| |
Collapse
|
11
|
Gutiérrez-Chamorro L, Felip E, Bernat-Peguera A, Ezeonwumelu IJ, Teruel I, Martínez-Cardús A, Clotet B, Riveira-Muñoz E, Romeo M, Margelí M, Ballana E. SAMHD1 expression modulates innate immune activation and correlates with ovarian cancer prognosis. Front Immunol 2023; 14:1112761. [PMID: 36845138 PMCID: PMC9948397 DOI: 10.3389/fimmu.2023.1112761] [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: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Purpose SAMHD1 is a deoxynucleotide triphosphate (dNTP) triphosphohydrolase which has been proposed as a putative prognostic factor in haematological cancers and certain solid tumours, although with controversial data. Here, we evaluate SAMHD1 function in ovarian cancer, both in vitro and in ovarian cancer patients. Methods SAMHD1 expression was downregulated in ovarian cancer cell lines OVCAR3 and SKOV3 by RNA interference. Gene and protein expression changes in immune signalling pathways were assessed. SAMHD1 expression in ovarian cancer patients was evaluated by immunohistochemistry and survival analysis was performed according to SAMHD1 expression. Results SAMHD1 knockdown induced a significant upregulation of proinflammatory cytokines concomitant to increased expression of the main RNA-sensors, MDA5 and RIG-I, and interferon-stimulated genes, supporting the idea that the absence of SAMHD1 promotes innate immune activation in vitro. To assess the contribution of SAMHD1 in ovarian cancer patients, tumours were stratified in SAMHD1-low and SAMHD1-high expressing tumours, resulting in significantly shorter progression free survival (PFS) and overall survival (OS) in SAMHD1-high expression subgroup (p=0.01 and 0.04, respectively). Conclusions SAMHD1 depletion correlates with increased innate immune cell signalling in ovarian cancer cells. In clinical samples, SAMHD1-low expressing tumors showed increased progression free survival and overall survival irrespective of BRCA mutation status. These results point towards SAMHD1 modulation as a new therapeutic strategy, able to enhance innate immune activation directly in tumour cells, leading to improved prognosis in ovarian cancer.
Collapse
Affiliation(s)
- Lucía Gutiérrez-Chamorro
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Eudald Felip
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain,Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Adrià Bernat-Peguera
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Ifeanyi Jude Ezeonwumelu
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Iris Teruel
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Anna Martínez-Cardús
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain,Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Riveira-Muñoz
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Margarita Romeo
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain,*Correspondence: Margarita Romeo, ; Mireia Margelí, ; Ester Ballana,
| | - Mireia Margelí
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO (Badalona Applied Research Group in Oncology), Health Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain,*Correspondence: Margarita Romeo, ; Mireia Margelí, ; Ester Ballana,
| | - Ester Ballana
- IrsiCaixa AIDS Research Institute – and Health Research Institute Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain,*Correspondence: Margarita Romeo, ; Mireia Margelí, ; Ester Ballana,
| |
Collapse
|
12
|
Ölmez F, Oğlak SC, Ölmez ÖF, Akbayır Ö, Yılmaz E, Akgöl S, Konal M, Seyhan NA, Kinter AK. High expression of CD8 in the tumor microenvironment is associated with PD-1 expression and patient survival in high-grade serous ovarian cancer. Turk J Obstet Gynecol 2022; 19:246-256. [PMID: 36149309 PMCID: PMC9511932 DOI: 10.4274/tjod.galenos.2022.59558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: The current study assesses programmed death-1 (PD-1) receptor expression and CD3, CD4, and CD8 tumor-infiltrating lymphocytes (TILs) in high-grade serous ovarian cancer (HGSOC) and associates our results with neoadjuvant chemotherapy history and disease prognosis. Materials and Methods: We included cases diagnosed with primary HGSOC with biopsy or surgical resection materials in this study. The immunoreactivity of CD3, CD4, CD8, and PD1 was assessed immunohistochemically in tumor tissue. We analyzed TILs in two predetermined groups of high and low TIL. The relationships between clinical characteristics, PD-1, and TIL were assessed. by the χ(2) test or Fisher’s Exact test. We used Kaplan-Meier survival analysis and Cox proportional hazards regression model to the connection between survival and the amounts of TIL, and PD1. Results: Univariate analysis demonstrated that optimal debulking (p<0.001), early International Federation of Gynecology and Obstetrics stage (p=0.046), and higher scores of stromal CD8+ TIL expression (p=0.028) in tumor cells were all substantially correlated with longer disease-free survival (DFS), whereas the remaining variables analyzed, including PD-1 positivity, stromal CD3+, and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs, were not correlated with DFS. Also, univariate analysis revealed that optimal debulking (p=0.010), and higher scores of stromal CD8+ TIL expression (p=0.021) in tumor cells were all substantially correlated with longer overall survival (OS). Conclusion: Higher scores of stromal CD8+ TILs are substantially correlated with DFS and OS in univariate analyses, whereas scores of stromal CD3+ and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs are not correlated with DFS and OS in both univariate and multivariate analyses. Also, we found a significant association between PD-1 positivity and the scores of stromal CD3+ TILs and intraepithelial CD8+ TILs. However, no remarkable relationship was revealed between PD-1 positivity and the survival of HGSOC cases.
Collapse
|
13
|
Peripheral lymphocyte populations in ovarian cancer patients and correlations with clinicopathological features. J Ovarian Res 2022; 15:43. [PMID: 35410290 PMCID: PMC8996636 DOI: 10.1186/s13048-022-00977-3] [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: 04/06/2021] [Accepted: 04/05/2022] [Indexed: 11/18/2022] Open
Abstract
Background To investigate the alterations of peripheral lymphocyte subpopulations in ovarian cancer patients compared to benign or borderline counterparts. The possible clinicopathological implications were also evaluated. Methods We enrolled 112 treatment-naive ovarian cancer patients, 14 borderline tumor patients and 44 benign tumor patients between 09/2016 and 01/2019. Flow cytometry was used to measure the peripheral lymphocyte subsets consisting of T cells (CD3+, CD3+CD4+, CD3+CD8+ and CD8+CD28+), regulatory T cells (Tregs, CD4+CD25+CD127−), natural killer cells (NK cells, CD3−CD56+) and B cells (CD19+). Results Most ovarian cancer patients were high-grade serous carcinoma (84.8%), followed by clear cell carcinoma (8.03%). Late-stage tumor (FIGO III + IV) accounted for 82.1%. The study showed that the proportions of peripheral lymphocyte subsets underwent apparent changes in ovarian cancer patients. We observed elevated levels of Treg cells in patients with both ovarian borderline and malignant tumor compared to those with benign tumors, which achieved statistic significance. In contrast, CD3+CD8+ T and CD8+CD28+ T cells were significantly lower in ovarian cancer patients. Interestingly, low level of B cells was correlated to clear cell carcinoma (P = 0.024), advanced tumor (P = 0.028) and platinum-resistant recurrence (P = 0.014). Regarding the changes of lymphocyte subsets after surgery, CD8+CD28+ T cells had a significant decreasing tendency (P = 0.007) while B cells were the opposite (P < 0.001). Conclusions Ovarian cancer patients have altered circulating lymphocyte profile (elevated Treg cell, depressed CD3+CD8+ T and CD8+CD28+ T cells). Low level of B cells might be related to disease aggressiveness, and it recovered after the removal of tumor, which merits further study. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-022-00977-3. 1 Ovarian cancer patients have altered circulating lymphocyte profile 2 Low level of B cells might be related to disease aggressiveness, and it recovered after the removal of tumor.
Collapse
|
14
|
Vyas D, Patel M, Wairkar S. Strategies for active tumor targeting-an update. Eur J Pharmacol 2022; 915:174512. [PMID: 34555395 DOI: 10.1016/j.ejphar.2021.174512] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 01/26/2023]
Abstract
A complete cure for cancer is still the holy grail for scientists. The existing treatment of cancer is primarily focused on surgery, radiation and conventional chemotherapy. However, chemotherapeutic agents also affect healthy tissues or organs due to a lack of specificity. While passive targeting is studied for anticancer drugs focused on the enhanced permeability and retention effect, it failed to achieve drug accumulation at the tumor site and desired therapeutic efficacy. This review presents an outline of the current significant targets for active tumor drug delivery systems and provides insight into the direction of active tumor-targeting strategies. For this purpose, a systematic understanding of the physiological factors, tumor microenvironment and its components, overexpressed receptor and associated proteins are covered here. We focused on angiogenesis mediated targeting, receptor-mediated targeting and peptide targeting. This active targeting along with integration with nano delivery systems helps in achieving specific action, thus reducing the associated adverse effects to healthy tissues. Although the tumor-targeting methods and possibilities explored so far seem revolutionary in cancer treatment, in-depth clinical studies data is required for its commercial translation.
Collapse
Affiliation(s)
- Darshan Vyas
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Mital Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
15
|
McNamara B, Guerra R, Qin J, Craig AD, Chen LM, Varma MG, Chapman JS. Survival impact of bowel resection at the time of interval cytoreductive surgery for advanced ovarian cancer. Gynecol Oncol Rep 2021; 38:100870. [PMID: 34646929 PMCID: PMC8496105 DOI: 10.1016/j.gore.2021.100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
Objectives To evaluate the impact of bowel resection at the time of interval cytoreductive surgery on survival. Methods We identified patients with advanced ovarian cancer who underwent neoadjuvant chemotherapy and interval cytoreductive surgery between 2008 and 2018 from a single-institution tumor registry. Kaplan-Meier survival analysis and Cox proportional hazards models were performed comparing patients who underwent bowel resection to those who did not. Results Of 158 patients, 43 (27%) underwent bowel resection. Rates of optimal (95%) and sub-optimal (5%) resection did not differ with bowel resection. Patients that required bowel resection had worse three-year survival (43% vs. 63%), even after adjusting for confounding variables of age, stage, number of neoadjuvant cycles, R0 resection, and ASA score (HR 2.27, p < 0.01). Adjusted progression-free survival did not differ between groups (HR 0.92, p = 0.72). Patients who underwent bowel resection were more likely to require blood transfusion (p < 0.01), and have a longer hospital stay (5 days vs 7.5 days, p < 0.01). Conclusions Bowel resection at the time of interval cytoreduction confers a greater than 2-fold increased risk of mortality and does not impact progression-free survival. Long-term sequelae of the peri-operative morbidity of bowel resection may contribute to increased mortality, and bowel resection may be a surrogate for disease biology with poor prognosis.
Collapse
Affiliation(s)
- Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Rosa Guerra
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer Qin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amaranta D Craig
- Department of Gynecologic Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Lee-May Chen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Madhulika G Varma
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jocelyn S Chapman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
16
|
Liao C, Wang A, Ma Y, Liu H. Long non-coding RNA FOXP4-AS1 is a prognostic biomarker and associated with immune infiltrates in ovarian serous cystadenocarcinoma. Medicine (Baltimore) 2021; 100:e27473. [PMID: 34622876 PMCID: PMC8500601 DOI: 10.1097/md.0000000000027473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND FOXP4-AS1 expression participates in multiple signal pathways and has been previously reported in colorectal cancer, cervical cancer, and other cancer cells. However, its role on prognosis and immune infiltrates in ovarian serous cystadenocarcinoma (OVs) remains unclear. The purpose of our study was to investigate the expression of FOXP4-AS1 in OVs and its association with immune infiltrates, and determined its prognostic roles in OVs. METHODS Using The Cancer Genome Atlas (TCGA) database, we retrieved FOXP4-AS1 expression and clinical information for 376 patients with OVs. Wilcoxon rank sum test was used to compare the expression of FOXP4-AS1 in OVs and normal ovarian tissue. Logistic regression was used to analyze the relationship between clinicopathologic features and FOXP4-AS1. Gene Set Enrichment Analysis (GSEA), and single sample Gene Set Enrichment Analysis (ssGSEA) was conducted to investigate the enrich pathways and functions and quantify the extent of immune cells infiltration for FOXP4-AS1. Kaplan-Meier method was used to generate survival curves, and Cox regression was used to analyze the relationship between FOXP4-AS1 and survival rate. RESULTS High FOXP4-AS1 expression was significantly correlated with tumor FIGO stage (P = .026). Multivariate survival analysis showed that FOXP4-AS1was an independent prognostic marker for overall survival (OS; hazard ratio [HR]: 0.638; 95% confidence interval [CI]:0.467-0.871; P = .001) and disease-specific survival (DSS; HR: 0.649; CI: 0.476-0.885; P = .006). GSEA showed that High FOXP4-AS1 expression may active programmed cell death 1 (PD-1) signaling, the cytotoxic T lymphocyte-associated antigen-4 (CTLA4) pathway, the B cell receptor signaling pathway, apoptosis, fibroblast growth factor receptor (FGFR) signaling, and the Janus-activated kinase signal transducers and activators of transcription (JAK-STAT) signaling pathway. FOXP4-AS1 expression was negatively correlated with markers of immune cells, including aDC, cytotoxic cells and neutrophils. CONCLUSION High FOXP4-AS1 expression has the potential to be a prognostic molecular marker of favorable survival in OVs.
Collapse
Affiliation(s)
- Cheng Liao
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Ao Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Yushan Ma
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| | - Hui Liu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P. R. China
| |
Collapse
|
17
|
Liu C, Barger CJ, Karpf AR. FOXM1: A Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer. Cancers (Basel) 2021; 13:3065. [PMID: 34205406 PMCID: PMC8235333 DOI: 10.3390/cancers13123065] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Forkhead box M1 (FOXM1) is a member of the conserved forkhead box (FOX) transcription factor family. Over the last two decades, FOXM1 has emerged as a multifunctional oncoprotein and a robust biomarker of poor prognosis in many human malignancies. In this review article, we address the current knowledge regarding the mechanisms of regulation and oncogenic functions of FOXM1, particularly in the context of ovarian cancer. FOXM1 and its associated oncogenic transcriptional signature are enriched in >85% of ovarian cancer cases and FOXM1 expression and activity can be enhanced by a plethora of genomic, transcriptional, post-transcriptional, and post-translational mechanisms. As a master transcriptional regulator, FOXM1 promotes critical oncogenic phenotypes in ovarian cancer, including: (1) cell proliferation, (2) invasion and metastasis, (3) chemotherapy resistance, (4) cancer stem cell (CSC) properties, (5) genomic instability, and (6) altered cellular metabolism. We additionally discuss the evidence for FOXM1 as a cancer biomarker, describe the rationale for FOXM1 as a cancer therapeutic target, and provide an overview of therapeutic strategies used to target FOXM1 for cancer treatment.
Collapse
Affiliation(s)
| | | | - Adam R. Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68918-6805, USA; (C.L.); (C.J.B.)
| |
Collapse
|
18
|
Liu S, Wu M, Wang F. Research Progress in Prognostic Factors and Biomarkers of Ovarian Cancer. J Cancer 2021; 12:3976-3996. [PMID: 34093804 PMCID: PMC8176232 DOI: 10.7150/jca.47695] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is a serious threat to women's health; its early diagnosis rate is low and prone to metastasis and recurrence. The current conventional treatment for ovarian cancer is a combination of platinum and paclitaxel chemotherapy based on surgery. The recurrence and progression of ovarian cancer with poor prognosis is a major challenge in treatment. With rapid advances in technology, understanding of the molecular pathways involved in ovarian cancer recurrence and progression has increased, biomarker-guided treatment options can greatly improve the prognosis of patients. This review systematically discusses and summarizes existing and new information on prognostic factors and biomarkers of ovarian cancer, which is expected to improve the clinical management of patients and lead to effective personalized treatment.
Collapse
Affiliation(s)
- Shuna Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Ming Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210029
- National Key Clinical Department of Laboratory Medicine, Nanjing, China, 210029
| |
Collapse
|
19
|
Baert T, Ferrero A, Sehouli J, O'Donnell DM, González-Martín A, Joly F, van der Velden J, Blecharz P, Tan DSP, Querleu D, Colombo N, du Bois A, Ledermann JA. The systemic treatment of recurrent ovarian cancer revisited. Ann Oncol 2021; 32:710-725. [PMID: 33675937 DOI: 10.1016/j.annonc.2021.02.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Treatment approaches for relapsed ovarian cancer have evolved over the past decade from a calendar-based decision tree to a patient-oriented biologically driven algorithm. Nowadays, platinum-based chemotherapy should be offered to all patients with a reasonable chance of responding to this therapy. The treatment-free interval for platinum is only one of many factors affecting patients' eligibility for platinum re-treatment. Bevacizumab increases the response to chemotherapy irrespective of the cytotoxic regimen and can be valuable in patients with an urgent need for symptom relief (e.g. pleural effusion, ascites). For patients with recurrent high-grade ovarian cancer, which responds to platinum-based treatment, maintenance therapy with a poly(ADP-ribose) polymerase inhibitor can be offered, regardless of the BRCA mutation status. Here we review contemporary decision-making processes in the systemic treatment of relapsed ovarian cancer.
Collapse
Affiliation(s)
- T Baert
- Department of Gynecology and Gynecological Oncology, Kliniken Essen-Mitte, Essen, Germany; Department of Oncology, KU Leuven, Leuven, Belgium.
| | - A Ferrero
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, Mauriziano Hospital, Turin, Italy
| | - J Sehouli
- Department of Gynecology with Center for Oncological Surgery, Charité-University hospital Berlin, Berlin, Germany
| | - D M O'Donnell
- Department of Oncology, St. James's Hospital, Dublin, Ireland
| | - A González-Martín
- Medical Oncology Department, Clínica Universidad de Navarra University Hospital, Madrid, Spain
| | - F Joly
- Department of Oncology, Centre Francois Baclesse, Caen, France
| | - J van der Velden
- Department of Medical Oncology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - P Blecharz
- Department of Gynecologic Oncology, Center of Oncology, M. Sklodowska-Curie Institute, Krakow, Poland
| | - D S P Tan
- Department of Haematology-Oncology, National University Cancer Institute of Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - D Querleu
- Department of Surgical Oncology, Institut Bergonié, Bordeaux, France
| | - N Colombo
- Department of Medicine and Surgery, European Institute of Oncology IRCCS, Milan, Italy; University of Milan-Bicocca, Milan, Italy
| | - A du Bois
- Department of Gynecology and Gynecological Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - J A Ledermann
- Cancer Research UK and UCL Cancer Trials Centre, UCL Cancer Institute, London, UK
| |
Collapse
|
20
|
Roane BM, Meza-Perez S, Katre AA, Goldsberry WN, Randall TD, Norian LA, Birrer MJ, Arend RC. Neutralization of TGFβ Improves Tumor Immunity and Reduces Tumor Progression in Ovarian Carcinoma. Mol Cancer Ther 2020; 20:602-611. [PMID: 33323456 DOI: 10.1158/1535-7163.mct-20-0412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/17/2020] [Accepted: 12/08/2020] [Indexed: 01/10/2023]
Abstract
The immunosuppressive effects of TGFβ promotes tumor progression and diminishes response to therapy. In this study, we used ID8-p53-/- tumors as a murine model of high-grade serous ovarian cancer. An mAb targeting all three TGFβ ligands was used to neutralize TGFβ. Ascites and omentum were collected and changes in T-cell response were measured using flow. Treatment with anti-TGFβ therapy every other day following injection of tumor cells resulted in decreased ascites volume (4.1 mL vs. 0.7 mL; P < 0.001) and improved the CD8:Treg ratio (0.37 vs. 2.5; P = 0.02) compared with untreated mice. A single dose of therapy prior to tumor challenge resulted in a similar reduction of ascites volume (2.7 vs. 0.67 mL; P = 0.002) and increased CD8:Tregs ratio (0.36 vs. 1.49; P = 0.007), while also significantly reducing omental weight (114.9 mg vs. 93.4 mg; P = 0.017). Beginning treatment before inoculation with tumor cells and continuing for 6 weeks, we observe similar changes and prolonged overall survival (median 70 days vs. 57.5 days). TGFβ neutralization results in favorable changes to the T-cell response within the tumor microenvironment, leading to decreased tumor progression in ovarian cancer. The utilization of anti-TGFβ therapy may be an option for management in patients with ovarian cancer to improve clinical outcomes and warrants further investigation.
Collapse
Affiliation(s)
- Brandon M Roane
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Selene Meza-Perez
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ashwini A Katre
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Whitney N Goldsberry
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama
| | - Lyse A Norian
- Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael J Birrer
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Rebecca C Arend
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama
| |
Collapse
|
21
|
den Ouden JE, Zaman GJ, Dylus J, van Doornmalen AM, Mulder WR, Grobben Y, van Riel WE, de Hullu JA, Buijsman RC, van Altena AM. Chemotherapy sensitivity testing on ovarian cancer cells isolated from malignant ascites. Oncotarget 2020; 11:4570-4581. [PMID: 33346216 PMCID: PMC7733621 DOI: 10.18632/oncotarget.27827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In epithelial ovarian cancer (EOC), 15-20% of the tumors do not respond to first-line chemotherapy (paclitaxel with platinum-based therapy), and in recurrences this number increases. Our aim is to determine the feasibility of cell proliferation assays of tumor cells isolated from malignant ascites to predict in vitro chemotherapy sensitivity, and to correlate these results with clinical outcome. MATERIALS AND METHODS Ascites was collected from twenty women with advanced EOC. Cell samples were enriched for tumor cells and EOC origin was confirmed by intracellular staining of CK7, surface staining of CA125 and EpCAM, and HE4 gene expression. In vitro sensitivity to chemotherapy was determined in cell proliferation assays using intracellular ATP content as an indirect measure of cell number. In vitro drug response was quantified by calculation of the drug concentration at which cell growth was inhibited with 50%. Clinical outcome was determined using post-treatment CA125 level. RESULTS Cell samples of twenty patients were collected, of which three samples that failed to proliferate were excluded in the analysis (15%). Three other samples were excluded, because clinical outcome could not be determined correctly. In twelve of the fourteen remaining cases (86%) in vitro drug sensitivity and clinical outcome corresponded, while in two samples (14%) there was no correspondence. CONCLUSIONS Our study demonstrates the feasibility of drug sensitivity tests using tumor cells isolated from ascites of advanced EOC patients. Larger observational studies are required to confirm the correlation between the in vitro sensitivity and clinical outcome.
Collapse
Affiliation(s)
- Judith E. den Ouden
- Radboud Institute for Health Sciences, Radboud University Medical Center, Obstetrics and Gynecology, Nijmegen, The Netherlands
| | - Guido J.R. Zaman
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| | - Jelle Dylus
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| | | | | | - Yvonne Grobben
- Netherlands Translational Research Center B.V., Oss, The Netherlands
| | | | - Joanne A. de Hullu
- Radboud Institute for Health Sciences, Radboud University Medical Center, Obstetrics and Gynecology, Nijmegen, The Netherlands
| | | | - Anne M. van Altena
- Radboud Institute for Health Sciences, Radboud University Medical Center, Obstetrics and Gynecology, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Jäntti T, Luhtala S, Mäenpää J, Staff S. Characterization of immunoreactivity with whole-slide imaging and digital analysis in high-grade serous ovarian cancer. Tumour Biol 2020; 42:1010428320971404. [PMID: 33169632 DOI: 10.1177/1010428320971404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer is the most lethal of gynecological cancers with 5-year survival rate of ca. 45%. The most common histologic subtype is high-grade serous carcinoma, which typically is presented with advanced stage and development of chemoresistance. Therefore, new treatment options, including immunotherapies, are needed. Understanding the features of the immune cell populations in the tumor microenvironment is essential for developing personalized treatments and finding predictive biomarkers. Digital image analysis may enhance the accuracy and reliability of immune cell infiltration assessment in the tumor microenvironment. The aim of this study was to characterize tumor microenvironment in a retrospective cohort of high-grade serous carcinoma samples with whole-slide imaging and digital image analysis. Formalin-fixed paraffin-embedded high-grade serous carcinoma tumor tissue samples (n = 67) were analyzed for six immunohistochemical stainings: CD4, CD8, FoxP3, granzyme B, CD68, and CD163. The stained sample slides were scanned into a digital format and assessed using QuPath 0.1.2 and ImageJ software. Staining patterns were associated with clinicopathological data. The higher numbers of intraepithelial CD8+, CD163+, and granzyme B+ immune cells were associated with survival benefit when analyzed individually, while high levels of both CD8+ and granzyme B+ tumor-infiltrating lymphocytes were an independent prognostic factor in the Cox multivariate regression analysis (median progression-free survival; hazard ratio = 0.287, p = 0.002). Specimens taken after administration of neoadjuvant chemotherapy presented with lower FoxP3+ tumor-infiltrating lymphocyte density (Fisher's exact test, p = 0.013). However, none of the studied immunomarkers was associated with overall survival or clinical factors. Tumors having high amount of both intraepithelial CD8+ and granzyme B+ tumor-infiltrating lymphocytes showed better progression-free survival, possibly reflecting an activated immune state in the tumor microenvironment. The combined positivity of CD8 and granzyme B warrants further investigation with respect to predicting response to immune therapy. Neoadjuvant chemotherapy may have an effect on the tumor microenvironment and therefore on the response to immuno-oncologic or chemotherapy treatments.
Collapse
Affiliation(s)
- Tiina Jäntti
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Satu Luhtala
- Department of Pathology, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Johanna Mäenpää
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland.,Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Synnöve Staff
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland.,Tays Cancer Centre, Tampere University Hospital, Tampere, Finland.,Department of Gynecology and Obstetrics, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
23
|
Winkler I, Woś J, Bojarska-Junak A, Semczuk A, Rechberger T, Baranowski W, Markut-Miotła E, Tabarkiewicz J, Wolińska E, Skrzypczak M. An association of iNKT+/CD3+/CD161+ lymphocytes in ovarian cancer tissue with CA125 serum concentration. Immunobiology 2020; 225:152010. [PMID: 33130518 DOI: 10.1016/j.imbio.2020.152010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/01/2022]
Abstract
The purpose of this study was to investigate the association of iNKT (human invariant natural killer T) cells with the key marker of ovarian cancer (OC) - CA125 (cancer antigen125) in serum. The study reports the assessment of iNKT cells in peripheral blood and tissue of benign and borderline ovarian tumors (BOTs) and in the advanced-stage ovarian cancer. The study groups were as follows: 25 women with benign ovarian tumors, 11 women with BOTs, and 24 women with primary advanced-stage ovarian cancers. The control group consisted of 20 patients without the ovarian pathology. The rates of iNKT lymphocytes in the peripheral blood and tissue specimens were evaluated by a flow cytometry. Significant differences in the percentage of iNKT+/CD3+ of CD3+ lymphocytes, iNKT+/CD3+/CD161+ among CD3+ and iNKT+/CD3+/CD161+ among CD3+/iNKT+ between the control group and patients with ovarian tumors in the peripheral blood and tumor tissue were identified. Significant correlations were noticed between the proportion of lymphocytes iNKT+/CD3+/CD161+ among CD3+/iNKT cells in blood and in cancer tissue of both benign and malignant tumors. In the OC group, neither the ratio of iNKT cells in the blood (P = 0.07), nor the intra-tumor NKT-cell infiltration (P = 0.5) were independent prognostic factors for the follow-up. An increased rate of iNKT cells was detected in benign ovarian tumors compared to OCs. In patients with ovarian cancer, a higher rate of iNKT cells in tumor tissue was present related to that noted in the patient's blood. In addition, a correlation was discovered between the CA125 serum marker and NKT cells from the ovarian cancer tissue. This article has for the first time demonstrated a negative relationship between serum levels and NKT lymphocyte count from ovarian tissue. The inflammatory process in ovarian cancer tissue and the potential infiltration of endothelial immune cells, may result in a reduced number of NKT cells in the tumor microenvironment and increased circulation of the CA125 marker. Presented findings underscore new aspects of the iNKT cells involvement in the ovarian cancer development.
Collapse
Affiliation(s)
- Izabela Winkler
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland; IInd Department of Gynecology, St' Johns Center Oncology, 7 Jaczewski Street, 20-090, Lublin, Poland.
| | - Justyna Woś
- Department of Clinical Immunology, Lublin Medical University, 4a Chodźki Street, 20-093, Lublin, Poland
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Lublin Medical University, 4a Chodźki Street, 20-093, Lublin, Poland
| | - Andrzej Semczuk
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| | - Tomasz Rechberger
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| | - Włodzimierz Baranowski
- IInd Department of Gynecology, St' Johns Center Oncology, 7 Jaczewski Street, 20-090, Lublin, Poland; Military Institute of Medicine, Department of Gynecology and Oncological Gynecology, 38 Szaserów street, Warsaw, Poland
| | - Ewa Markut-Miotła
- Department of Pediatric Pulmonology and Rheumatology, Lublin Medical University, 8 Jaczewski Street, 20-090, Lublin, Poland
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, 1A Warzywna Street, 35-959 Rzeszów, Poland
| | - Ewa Wolińska
- Department of Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Skrzypczak
- IInd Department of Gynecology, Lublin Medical University, 8 Jaczewski Street, 20-954, Lublin, Poland
| |
Collapse
|
24
|
Ren XY, Yang WB, Tian Y. Overexpression of long noncoding RNA PTPRG-AS1 is associated with poor prognosis in epithelial ovarian cancer. ACTA ACUST UNITED AC 2020; 66:948-953. [PMID: 32844927 DOI: 10.1590/1806-9282.66.7.948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 02/26/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) have been shown to play a critical role in tumor progression. Abnormal expression of LncRNA PTPRG antisense RNA 1 (PTPRG-AS1) has been reported in several tumors. Hence, we aimed to determine the expression and clinical significance of PTPRG-AS1 in epithelial ovarian cancer (EOC) patients. METHODS The expressions of PTPRG-AS1 were assessed in 184 pairs of EOC tumor specimens and adjacent normal tissues. The levels of target lncRNAs and GAPDH were examined using standard SYBR-Green methods. The relationships between the expressions of PTPRG-AS1 and the clinicopathological features were analyzed using the chi-square test. Multivariate analysis using the Cox proportional hazards model was performed to assess the prognostic value of PTPRG-AS1 in EOC patients. RESULTS We confirmed that the expressions of PTPRG-AS1 were distinctly higher in the EOC tissue compared with the adjacent non-tumor specimens (p < 0.01). Higher levels of PTPRG-AS1 in EOC patients were associated with advanced FIGO stage (p = 0.005), grade (p = 0.006), and distant metastasis (p = 0.005). Survival analyses revealed that patients with high expressions of PTPRG-AS1 had a distinctly decreased overall survival (p = 0.0029) and disease-free survival (p = 0.0009) compared with those with low expressions of PTPRG-AS1. Multivariate assays indicated that PTPRG-AS1 expression was an independent prognostic factor for both overall survival and disease-free survival in EOC (Both p < 0.05). CONCLUSIONS Our study suggests that PTPRG-AS1 may serve as a novel prognostic biomarker for EOC patients.
Collapse
Affiliation(s)
- Xue-Ying Ren
- Department of Gynecology, Heji Hospital Affiliated, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wei-Bin Yang
- Heping Hospital Affiliated, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yun Tian
- Teaching and Research Office of Embryology, Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
25
|
Asare-Werehene M, Communal L, Carmona E, Han Y, Song YS, Burger D, Mes-Masson AM, Tsang BK. Plasma Gelsolin Inhibits CD8 + T-cell Function and Regulates Glutathione Production to Confer Chemoresistance in Ovarian Cancer. Cancer Res 2020; 80:3959-3971. [PMID: 32641415 DOI: 10.1158/0008-5472.can-20-0788] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/28/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Although initial treatment of ovarian cancer is successful, tumors typically relapse and become resistant to treatment. Because of poor infiltration of effector T cells, patients are mostly unresponsive to immunotherapy. Plasma gelsolin (pGSN) is transported by exosomes (small extracellular vesicle, sEV) and plays a key role in ovarian cancer chemoresistance, yet little is known about its role in immunosurveillance. Here, we report the immunomodulatory roles of sEV-pGSN in ovarian cancer chemoresistance. In chemosensitive conditions, secretion of sEV-pGSN was low, allowing for optimal CD8+ T-cell function. This resulted in increased T-cell secretion of IFNγ, which reduced intracellular glutathione (GSH) production and sensitized chemosensitive cells to cis-diaminedichloroplatinum (CDDP)-induced apoptosis. In chemoresistant conditions, increased secretion of sEV-pGSN by ovarian cancer cells induced apoptosis in CD8+ T cells. IFNγ secretion was therefore reduced, resulting in high GSH production and resistance to CDDP-induced death in ovarian cancer cells. These findings support our hypothesis that sEV-pGSN attenuates immunosurveillance and regulates GSH biosynthesis, a phenomenon that contributes to chemoresistance in ovarian cancer. SIGNIFICANCE: These findings provide new insight into pGSN-mediated immune cell dysfunction in ovarian cancer chemoresistance and demonstrate how this dysfunction can be exploited to enhance immunotherapy.
Collapse
Affiliation(s)
- Meshach Asare-Werehene
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Laudine Communal
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Euridice Carmona
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Youngjin Han
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Sang Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Dylan Burger
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du CHUM et Institut du Cancer de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Benjamin K Tsang
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada. .,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
26
|
Abstract
Engagement of activating receptor NKG2D to its ligand mediates natural killer (NK) cell activation and enhances cytotoxicity. NKG2D ligands (NKG2DLs) are frequently expressed on the tumor cell surface. However, the expression patterns of different NKG2DLs vary between tumor cells. Downregulation of certain ligand enables the tumor cells to escape NK cell-mediated immunosurveillance. By generating tumor cell lines with high expression of NKG2D ligand MULT1, we aimed to explore the function of NKG2DLs diversity on the activation and regulation of NKG2D signaling pathway. NK cells were potently activated by the "acquired" MULT1 expression on MOVCAR 5009 cells. Further, the progression of the tumor was significantly inhibited in mice inoculated with MULT1-expressing MOVCAR 5009 cells. Also, the pulmonary metastasis of MULT1-expressing B16-F0 cells was also significantly reduced in vivo. Our results implied that "acquired" NKG2D ligands enhance antitumor responses of NK cells, providing insights for designing novel therapeutic strategies and drugs to enhance NK cell surveillance over malignances.
Collapse
|
27
|
Mlynska A, Vaišnorė R, Rafanavičius V, Jocys S, Janeiko J, Petrauskytė M, Bijeikis S, Cimmperman P, Intaitė B, Žilionytė K, Barakauskienė A, Meškauskas R, Paberalė E, Pašukonienė V. A gene signature for immune subtyping of desert, excluded, and inflamed ovarian tumors. Am J Reprod Immunol 2020; 84:e13244. [PMID: 32294293 DOI: 10.1111/aji.13244] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/24/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
PROBLEM The current tumor immunology paradigm emphasizes the role of the immune tumor microenvironment and distinguishes several histologically and transcriptionally different immune tumor subtypes. However, the experimental validation of such classification is so far limited to selected cancer types. Here, we aimed to explore the existence of inflamed, excluded, and desert immune subtypes in ovarian cancer, as well as investigate their association with the disease outcome. METHOD OF STUDY We used the publicly available ovarian cancer dataset from The Cancer Genome Atlas for developing subtype assignment algorithm, which was next verified in a cohort of 32 real-world patients of a known tumor subtype. RESULTS Using clinical and gene expression data of 489 ovarian cancer patients in the publicly available dataset, we identified three transcriptionally distinct clusters, representing inflamed, excluded, and desert subtypes. We developed a two-step subtyping algorithm with COL5A2 serving as a marker for separating excluded tumors, and CD2, TAP1, and ICOS for distinguishing between inflamed and desert tumors. The accuracy of gene expression-based subtyping algorithm in a real-world cohort was 75%. Additionally, we confirmed that patients bearing inflamed tumors are more likely to survive longer. CONCLUSION Our results highlight the presence of transcriptionally and histologically distinct immune subtypes among ovarian tumors and emphasize the potential benefit of immune subtyping as a clinical tool for treatment tailoring.
Collapse
Affiliation(s)
| | | | | | - Simonas Jocys
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | - Julija Janeiko
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | - Simas Bijeikis
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | | | | | - Aušrinė Barakauskienė
- Vilnius University, Vilnius, Lithuania.,Ltd Patologijos Diagnostika, Vilnius, Lithuania
| | | | | | | |
Collapse
|
28
|
Marinelli O, Annibali D, Aguzzi C, Tuyaerts S, Amant F, Morelli MB, Santoni G, Amantini C, Maggi F, Nabissi M. The Controversial Role of PD-1 and Its Ligands in Gynecological Malignancies. Front Oncol 2019; 9:1073. [PMID: 31681606 PMCID: PMC6803534 DOI: 10.3389/fonc.2019.01073] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
The programmed death-1 (PD-1, CD279) receptor with its ligands, programmed death ligand 1 (PD-L1, CD274, B7-H1), and programmed death ligand 2 (PD-L2, CD273, B7-DC), are the key players of one of the immune checkpoint pathways inhibiting T-cell activation. PD-L1 and PD-L2 are expressed in different cancer cells and their microenvironment, including infiltrating immune cells. However, their prognostic value is still debated and their role in the tumor microenvironment has not been fully elucidated yet. Considering the importance that cancer immunotherapy with anti-PD-1 and anti-PD-L1 antibodies gained in several tumor types, in this review article we aim to discuss the role of the PD-1/PD-L1/PD-L2 axis in gynecological cancers. PD-1 ligands have been detected in ovarian, cervical, vulvar and uterine cancers, and correlation with prognosis seems dependent from their distribution. About PD-L2, very few reports are available so far in gynecological malignancies, and its role is still not completely understood. Clinical trials using anti-PD-1 or anti-PD-L1 antibodies, but not anti-PD-L2, are currently ongoing, in all types of gynecological cancers. They have shown good safety profiles in a certain cohort of patients, but response rates remain low and many aspects remain controversial. In this review, we propose possible solutions to enhance the clinical efficacy of PD-1 axis targeting therapies. Regarding PD-L2, it might be useful to better clarify its role in order to improve the efficiency of immunotherapy in female malignancies.
Collapse
Affiliation(s)
- Oliviero Marinelli
- School of Pharmacy, University of Camerino, Camerino, Italy.,School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Daniela Annibali
- Gynecological Oncology, Oncology Department, LKI Leuven Cancer Institute KU Leuven-University of Leuven, Leuven, Belgium
| | | | - Sandra Tuyaerts
- Gynecological Oncology, Oncology Department, LKI Leuven Cancer Institute KU Leuven-University of Leuven, Leuven, Belgium
| | - Frédéric Amant
- Gynecological Oncology, Oncology Department, LKI Leuven Cancer Institute KU Leuven-University of Leuven, Leuven, Belgium.,Centre for Gynecologic Oncology Amsterdam (CGOA), Antoni Van Leeuwenhoek-Netherlands Cancer Institute (AvL-NKI), University Medical Center (UMC), Amsterdam, Netherlands
| | - Maria Beatrice Morelli
- School of Pharmacy, University of Camerino, Camerino, Italy.,School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Consuelo Amantini
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Federica Maggi
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | |
Collapse
|
29
|
De La Motte Rouge T, Corné J, Cauchois A, Le Boulch M, Poupon C, Henno S, Rioux-Leclercq N, Le Pabic E, Laviolle B, Catros V, Levêque J, Fautrel A, Le Gallo M, Legembre P, Lavoué V. Serum CD95L Level Correlates with Tumor Immune Infiltration and Is a Positive Prognostic Marker for Advanced High-Grade Serous Ovarian Cancer. Mol Cancer Res 2019; 17:2537-2548. [DOI: 10.1158/1541-7786.mcr-19-0449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/15/2019] [Accepted: 09/10/2019] [Indexed: 11/16/2022]
|
30
|
Abstract
For more than 20 years, the combination of a platinum agent and a taxane has served as the primary chemotherapy strategy for epithelial ovarian cancer. Alternative approaches employing these drugs (intraperitoneal drug delivery, neoadjuvant therapy) have favorably impacted outcomes in selected patient populations. Multiple single agent and combination therapy strategies have been delivered in the second-line (or later) setting with the goal to prolong survival and optimize quality-of-life. The anti-angiogenic agent bevacizumab has been shown to be clinically active when added to chemotherapy and delivered as a "maintenance" strategy in the front-line, platinum-sensitive recurrent and platinum-resistant settings. Several poly-(ADP-ribose) polymerase (PARP) inhibitors are currently utilized as single-agent "second-line" treatment options or in the maintenance setting. Recent clinical research efforts in ovarian cancer have focused on the potential role of checkpoint inhibitors used alone or in combination with PARP inhibitors or anti-angiogenic agents.
Collapse
|
31
|
Tian H, Hou L, Xiong Y, Cheng Q, Huang J. Effect of Dexmedetomidine-Mediated Insulin-Like Growth Factor 2 (IGF2) Signal Pathway on Immune Function and Invasion and Migration of Cancer Cells in Rats with Ovarian Cancer. Med Sci Monit 2019; 25:4655-4664. [PMID: 31230061 PMCID: PMC6604677 DOI: 10.12659/msm.915503] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background The aim of this study was to explore the effect of dexmedetomidine (DEX)-mediated insulin-like growth factor 2 (IGF2) signal pathway on immune function and cancer cell invasion and migration in rats with ovarian cancer. Material/Methods Forty rats with ovarian cancer were divided into 4 groups: model group, and low dose (0.2 μg/kg/hour DEX), medium dose (1.0 μg/kg/hour DEX), and high dose (5.0 μg/kg/hour DEX) DEX groups. In addition, 10 Fischer344 rats were selected as a normal group. Human NUTU-19 poorly differentiated epithelial ovarian cancer cell line cells were divided into 4 groups: a blank group and low dose, medium dose, and high dose DEX NUTU-19 groups. Results Compared with the normal group, in the other groups the serum interleukin (IL)-2 and interferon gamma (INF-γ) levels, CD4+ and CD8+ percentages, CD4+/CD8+ ratio, and transformation rate of splenic lymphocytes were decreased, and the serum tumor necrosis factor alpha (TNF-α) level, IGF2, insulin-like growth factor 1 receptor (IGF1R), insulin receptor substrate 1 (IRS1) mRNA, and protein expressions in ovarian tissue were increased (all P<0.05). Results in the DEX groups compared with model group were the opposite of those in the other groups compared with normal group (all P<0.05). Compared with the blank group, in the other groups the proliferation, invasion, and migration of ovarian cancer cells were reduced significantly (all P<0.05). Compared with the low dose DEX NUTU-19 group, in the high dose DEX NUTU-19 group the invasion and migration of ovarian cancer cells weakened significantly (both P<0.05). Conclusions A certain dose of DEX can effectively inhibit IGF2 signal pathway activation to improve the immune function of rats with ovarian cancer, inhibiting the invasion and migration of ovarian cancer cells.
Collapse
Affiliation(s)
- Hang Tian
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Lei Hou
- Department of Anesthesiology, Shanxi Provincial Cancer Hospital, Taiyuan, Shanxi, China (mainland)
| | - Yumei Xiong
- Department of Pediatric Emergency, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Qiuju Cheng
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| | - Junking Huang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
32
|
Baert T, Vankerckhoven A, Riva M, Van Hoylandt A, Thirion G, Holger G, Mathivet T, Vergote I, Coosemans A. Myeloid Derived Suppressor Cells: Key Drivers of Immunosuppression in Ovarian Cancer. Front Immunol 2019; 10:1273. [PMID: 31214202 PMCID: PMC6558014 DOI: 10.3389/fimmu.2019.01273] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/20/2019] [Indexed: 12/27/2022] Open
Abstract
The presence of tumor infiltrating lymphocytes (TILs) is associated with a longer overall survival in advanced stage epithelial ovarian cancer. Despite the prognostic impact of TILs, response to checkpoint-inhibitors and antigen-specific active immunotherapy is limited in ovarian cancer. The goal of our study was to investigate the interaction between ovarian cancer and the innate and adaptive immune system in the ID8-fLuc syngeneic ovarian cancer mouse model. For the in vivo experiments C57BL/6, B6.129S7-Rag1tm1Mom/J, and B6.129P2(SJL)-Myd88tm1.1Defr/J mice were inoculated with ID8-fLuc. In vivo depletion experiments were performed using clodronate liposomes (CL), anti-CD8a, anti-GR1, anti-colony stimulating factor 1 (anti-CSF1), and TMβ1 (anti-CD122). Immune read out was performed by fluorescent activated cell sorting analysis for effector T cells, regulatory T cells, natural killer cells, B cells, macrophages, and myeloid derived suppressor cells (MDSC), immunohistochemistry for MDSC and tumor-associated macrophages (TAM) and immunofluorescence for M1 and M2 TAM in the vascular context. The effect of MDSC on T cell proliferation and phenotype were studied in vitro. We discovered that the absence of T and B cells did not influence tumor growth or survival of B6.129S7-Rag1tm1Mom/J mice compared to immunocompetent C57BL/6 mice. CL-induced macrophage depletion promoted tumor proliferation and shortened survival in C57BL/6 mice (p = 0.004) and in B6.129S7-Rag1tm1Mom/J mice (p = 0.0005). During CL treatment, we observed a clear increase of pro-inflammatory cytokines (p ≤ 0.02) and monocytic MDSC (p ≤ 0.01). Selective depletion of MDSC by anti-GR1 improved survival, certainly in comparison to mice treated with anti-CSF1 (p = 0.01-median survival 91 vs. 67.5 days). B6.129P2(SJL)-Myd88tm1.1Defr/J mice displayed to a longer median survival compared to C57BL/6 mice (90 vs. 76 days). MDSC activated by ID8-fLuc conditioned medium or ascites of tumor-bearing mice showed T cell suppressive functions in vitro. Based on these findings, we conclude that the adaptive immune system does not efficiently control tumor growth in the ID8-fLuc model. In addition, we discovered a prominent role for MDSC as the driver of immunosuppression in the ID8-fLuc ovarian cancer mouse model.
Collapse
Affiliation(s)
- Thaïs Baert
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynecology and Gynecologic Oncology, Kliniken Essen Mitte, Essen, Germany
| | - Ann Vankerckhoven
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Anaïs Van Hoylandt
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gitte Thirion
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gerhardt Holger
- Vascular Patterning Lab, Center for Cancer Biology, VIB, KU Leuven, Leuven, Belgium
| | - Thomas Mathivet
- Vascular Patterning Lab, Center for Cancer Biology, VIB, KU Leuven, Leuven, Belgium.,PARCC, HEGP Institute (team 9), INSERM U970, Université Paris Descartes, Paris, France
| | - Ignace Vergote
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Gynecologic Oncology, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - An Coosemans
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| |
Collapse
|
33
|
Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A, Chiappinelli KB. Epigenetic therapy for ovarian cancer: promise and progress. Clin Epigenetics 2019; 11:7. [PMID: 30646939 PMCID: PMC6334391 DOI: 10.1186/s13148-018-0602-0] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is the deadliest gynecologic malignancy, with a 5-year survival rate of approximately 47%, a number that has remained constant over the past two decades. Early diagnosis improves survival, but unfortunately only 15% of ovarian cancers are diagnosed at an early or localized stage. Most ovarian cancers are epithelial in origin and treatment prioritizes surgery and cytoreduction followed by cytotoxic platinum and taxane chemotherapy. While most tumors will initially respond to this treatment, recurrence is likely to occur within a median of 16 months for patients who present with advanced stage disease. New treatment options separate from traditional chemotherapy that take advantage of advances in understanding of the pathophysiology of ovarian cancer are needed to improve outcomes. Recent work has shown that mutations in genes encoding epigenetic regulators are mutated in ovarian cancer, driving tumorigenesis and resistance to treatment. Several of these epigenetic modifiers have emerged as promising drug targets for ovarian cancer therapy. In this article, we delineate epigenetic abnormalities in ovarian cancer, discuss key scientific advances using epigenetic therapies in preclinical ovarian cancer models, and review ongoing clinical trials utilizing epigenetic therapies in ovarian cancer.
Collapse
Affiliation(s)
- Sara Moufarrij
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Monica Dandapani
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Elisa Arthofer
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Stephanie Gomez
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Aneil Srivastava
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Micael Lopez-Acevedo
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Alejandro Villagra
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Katherine B. Chiappinelli
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| |
Collapse
|
34
|
Uppendahl LD, Felices M, Bendzick L, Ryan C, Kodal B, Hinderlie P, Boylan KLM, Skubitz APN, Miller JS, Geller MA. Cytokine-induced memory-like natural killer cells have enhanced function, proliferation, and in vivo expansion against ovarian cancer cells. Gynecol Oncol 2019; 153:149-157. [PMID: 30658847 DOI: 10.1016/j.ygyno.2019.01.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/12/2018] [Accepted: 01/03/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Natural killer (NK) cells are lymphocytes well suited for adoptive immunotherapy. Attempts with adoptive NK cell immunotherapy against ovarian cancer have proven unsuccessful, with the main limitations including failure to expand and diminished effector function. We investigated if incubation of NK cells with interleukin (IL)-12, IL-15, and IL-18 for 16h could produce cytokine-induced memory-like (CIML) NK cells capable of enhanced function against ovarian cancer. METHODS NK cells were preactivated briefly with IL-12, IL-15, and IL-18, rested, then placed against ovarian cancer targets to assess phenotype and function via flow cytometry. Real-time NK-cell-mediated tumor-killing was evaluated. Using ascites cells and cell-free ascites fluid, NK cell proliferation and function within the immunosuppressive microenvironment was evaluated in vitro. Finally, CIML NK cells were injected intraperitoneal (IP) into an in vivo xenogeneic mouse model of ovarian cancer. RESULTS CIML NK cells demonstrate enhanced cytokine (IFN-γ) production and NK-cell-mediated killing of ovarian cancer. NK cells treated overnight with cytokines led to robust activation characterized by temporal shedding of CD16, induction of CD25, and enhanced proliferation. CIML NK cells proliferate more with enhanced effector function compared to controls in an immunosuppressive microenvironment. Finally, human CIML NK cells exhibited potent antitumor effects within a xenogeneic mouse model of ovarian cancer. CONCLUSIONS CIML NK cells have enhanced functionality and persistence against ovarian cancer in vitro and in vivo, even when exposed to ascites fluid. These findings provide a strategy for NK cell-based immunotherapy to circumvent the immunosuppressive nature of ovarian cancer.
Collapse
Affiliation(s)
- Locke D Uppendahl
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Martin Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Laura Bendzick
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Caitlin Ryan
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Behiye Kodal
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Peter Hinderlie
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Kristin L M Boylan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Amy P N Skubitz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Melissa A Geller
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
35
|
Roelofsen T, Wefers C, Gorris MAJ, Textor JC, Massuger LFAG, de Vries IJM, van Altena AM. Spontaneous Regression of Ovarian Carcinoma After Septic Peritonitis; A Unique Case Report. Front Oncol 2018; 8:562. [PMID: 30555799 PMCID: PMC6281979 DOI: 10.3389/fonc.2018.00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/12/2018] [Indexed: 11/23/2022] Open
Abstract
Despite advances in therapy, ovarian cancer remains the most lethal gynecological malignancy and prognosis has not substantially improved over the past 3 decades. Immunotherapy is a promising new treatment option. However, the immunosuppressive cancer microenvironment must be overcome for immunotherapy to be successful. Here, we present a unique case of spontaneous regression of ovarian carcinoma after septic peritonitis. A 79-year-old woman was diagnosed with stage IIIc ovarian cancer. The omental cake biopsy was complicated by sepsis. Although the patient recovered, her physical condition did not allow further treatment for her ovarian cancer. After 6 months, spontaneous regression of the tumor was observed during surgery. Analysis of the immune infiltrate in the tissues showed a shift from a pro-tumorigenic to an anti-tumorigenic immune response after sepsis. Strong activation of the immune system during sepsis overruled the immunosuppressive tumor microenvironment and allowed for a potent anti-tumor immune response. More understanding of immunological responses in cases with cancer and septic peritonitis might be crucial to identify potential new targets for immunotherapy.
Collapse
Affiliation(s)
- Thijs Roelofsen
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Christina Wefers
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Johannes C. Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Leon F. A. G. Massuger
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical CentreNijmegen, Netherlands
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
36
|
Chen Q, Zhang H. Smac combined with DDP can inhibit drug resistance of ovarian cancer through regulation of Survivin expression. Cancer Biomark 2018; 22:1-6. [PMID: 29562492 DOI: 10.3233/cbm-170325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ovarian cancer has the highest mortality rate among gynecological malignancies, presenting a major threat to women's life and health. It is essential to study the mechanisms of drug resistance to chemotherapy to identify ways to enhance drug-sensitivity. In recent years, many studies have shown that Smac/DIABLO is closely related to tumor drug resistance. Smac/DIABLO expression is markedly different between drug-resistant and chemo sensitive tumor cells. Up-regulation of Smac/DIABLO has been shown to increase tumor cell chemotherapy sensitivity. We found that Smac, combined with DDP greatly inhibited proliferation of subcutaneous xenografts of ovarian cancer cell line SKOV3/DDP without side effects. Mechanistic studies showed that Smac can inhibit the expression of Survivin, promote cell apoptosis of drug-resistant ovarian cancer cells and reverse the drug resistance.
Collapse
|
37
|
Pinto MP, Balmaceda C, Bravo ML, Kato S, Villarroel A, Owen GI, Roa JC, Cuello MA, Ibañez C. Patient inflammatory status and CD4+/CD8+ intraepithelial tumor lymphocyte infiltration are predictors of outcomes in high-grade serous ovarian cancer. Gynecol Oncol 2018; 151:10-17. [PMID: 30078505 DOI: 10.1016/j.ygyno.2018.07.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is the most prevalent and aggressive histologic type of ovarian cancer. To date, there are no reliable biomarkers to effectively predict patient prognosis. Studies have demonstrated inflammation and tumor infiltrating lymphocytes (TILs) correlate with a bad and good prognosis, respectively. Here, we sought to evaluate systemic inflammation and TILs as early prognostic markers of survival. METHODS Neutrophil-to-lymphocyte ratio (NLR) and serum Lactate Dehydrogenase (LDH) were used as indicators of systemic inflammation. NLR, serum LDH, tumor infiltrating lymphocytes (TILs), PDL1 and quality of debulking surgery were evaluated as determinants of progression-free survival (PFS) and overall survival (OS) in a cohort of 128 HGSOC patients. RESULTS Initial univariate analysis showed that systemic inflammation measures (NLR and serum LDH), debulking surgery, and intra-epithelial TILs have a significant impact on both PFS and OS. After adjustment for several variables, multivariate analyses confirmed intraepithelial CD4+ T-cells, systemic inflammation measures, PDL1 and debulking surgery as determinants of better OS and PFS. CONCLUSIONS Systemic inflammation and TILs are early determinants of OS in HGSOC. Other variables such as the quality of debulking surgery and PDL1 also improve survival of patients. Regarding TIL sub-populations, intraepithelial CD4+ cells are associated to an increase in both PFS and OS. We also confirmed previous reports that demonstrate intraepithelial CD8+ cells correlate with an increase on PFS in ovarian cancer. A combined score using systemic inflammation and TILs may be of prognostic value for HGSOC patients.
Collapse
Affiliation(s)
- Mauricio P Pinto
- Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Carlos Balmaceda
- Unidad de Evaluación de Tecnologías Sanitarias. Centro de Investigación Clínica, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Chile
| | - Maria L Bravo
- Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; Millennium Institute for Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Chile
| | - Sumie Kato
- Departamento de Ginecología-Obstetricia, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Alejandra Villarroel
- Departamento de Anatomía Patológica, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Gareth I Owen
- Millennium Institute for Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Chile; Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Juan Carlos Roa
- Departamento de Anatomía Patológica, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Mauricio A Cuello
- Departamento de Ginecología-Obstetricia, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Carolina Ibañez
- Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; Millennium Institute for Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Chile; Centro del Cáncer UC-Christus, Chile.
| |
Collapse
|
38
|
Anti-CD73 and anti-OX40 immunotherapy coupled with a novel biocompatible enzyme prodrug system for the treatment of recurrent, metastatic ovarian cancer. Cancer Lett 2018; 425:174-182. [PMID: 29574275 DOI: 10.1016/j.canlet.2018.03.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 02/02/2023]
Abstract
Approximately 75% of ovarian cancer is diagnosed once metastasis to the peritoneal cavity has occurred. A large proportion of patients eventually develop platinum-resistive tumors, which are considered terminal. In order to provide an alternative a novel fusion protein, mCTH-ANXA5, has been developed for the treatment of recurrent, metastatic ovarian cancer. The fusion protein combines annexin V (ANXA5), an ovarian tumor and tumor vasculature targeting protein, with mutated cystathionine gamma-lyase (mCTH), an enzyme that converts selenomethionine (SeMet) into toxic methylselenol, which generates reactive oxygen species and eventual tumor cell death. In order to further enhance the therapeutic efficacy, anti-CD73 and anti-OX40 immunostimulants were combined with mCTH-ANXA5, resulting in an increase of survival by 100% from 12 to 24 days post-therapy and decrease tumor burden in mice with orthotopic metastatic ovarian cancer. Further evaluation of the combination therapy revealed a strong antibody-mediated immune response, and an increased infiltration of cytotoxic T-cells along with a decrease in tumor promoting immune cells. This study demonstrates the efficacy of a synergistic, multi-drug system by attacking the tumor as well as enlisting the body's own defense system to treat the patient.
Collapse
|
39
|
Stanske M, Wienert S, Castillo-Tong DC, Kreuzinger C, Vergote I, Lambrechts S, Gabra H, Gourley C, Ganapathi RN, Kolaschinski I, Budczies J, Sehouli J, Ruscito I, Denkert C, Kulbe H, Schmitt W, Jöhrens K, Braicu I, Darb-Esfahani S. Dynamics of the Intratumoral Immune Response during Progression of High-Grade Serous Ovarian Cancer. Neoplasia 2018; 20:280-288. [PMID: 29466768 PMCID: PMC5852388 DOI: 10.1016/j.neo.2018.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TILs) have an established impact on the prognosis of high-grade serous ovarian carcinoma (HGSOC), however, their role in recurrent ovarian cancer is largely unknown. We therefore systematically investigated TIL densities and MHC class I and II (MHC1, 2) expression in the progression of HGSOC. EXPERIMENTAL DESIGN CD3+, CD4+, CD8+ TILs and MHC1, 2 expression were evaluated by immunohistochemistry on tissue microarrays in 113 paired primary and recurrent HGSOC. TILs were quantified by image analysis. All patients had been included to the EU-funded OCTIPS FP7 project. RESULTS CD3+, CD4+, CD8+ TILs and MHC1 and MHC2 expression showed significant correlations between primary and recurrent tumor levels (Spearman rho 0.427, 0.533, 0.361, 0.456, 0.526 respectively; P<.0001 each). Paired testing revealed higher CD4+ densities and MHC1 expression in recurrent tumors (Wilcoxon P=.034 and P=.018). There was also a shift towards higher CD3+ TILs levels in recurrent carcinomas when analyzing platinum-sensitive tumors only (Wilcoxon P=.026) and in pairs with recurrent tumor tissue from first relapse only (Wilcoxon P=.031). High MHC2 expression was the only parameter to be significantly linked to prolonged progression-free survival after first relapse (PFS2, log-rank P=.012). CONCLUSIONS This is the first study that analyzed the development of TILs density and MHC expression in paired primary and recurrent HGSOC. The level of the antitumoral immune response in recurrent tumors was clearly dependent on the one in the primary tumor. Our data contribute to the understanding of temporal heterogeneity of HGSOC immune microenvironment and have implications for selection of samples for biomarker testing in the setting of immune-targeting therapeutics.
Collapse
Affiliation(s)
- Mandy Stanske
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Stephan Wienert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany; VM Scope GmbH, Charitéplatz 1, 10117 Berlin, Germany.
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | - Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | - Ignace Vergote
- Department of Gynecology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | | | - Hani Gabra
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, MRC IGMM, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK.
| | - Ram N Ganapathi
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas Health Care System, 1021 Morehead Medical Drive, Charlotte, NC 28204-2839, USA.
| | - Ivonne Kolaschinski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jan Budczies
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jalid Sehouli
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Ilary Ruscito
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany; UP Cell Therapy and Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy.
| | - Carsten Denkert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Hagen Kulbe
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Wolfgang Schmitt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Korinna Jöhrens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Ioana Braicu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology, Augustenburger Platz 1, 13353 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Silvia Darb-Esfahani
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Charitéplatz 1, 10117 Berlin, Germany; Tumorbank Ovarian Cancer Network (TOC), Department of Gynecology, Charité University Hospital Berlin, Germany, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
40
|
Zhang J, Ming C, Zhang W, Okechukwu PN, Morak-Młodawska B, Pluta K, Jeleń M, Akim AM, Ang KP, Ooi KK. 10 H-3,6-Diazaphenothiazine induces G 2/M phase cell cycle arrest and caspase-dependent apoptosis and inhibits cell invasion of A2780 ovarian carcinoma cells through the regulation of NF-κB and (BIRC6-XIAP) complexes. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3045-3063. [PMID: 29123378 PMCID: PMC5661483 DOI: 10.2147/dddt.s144415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The asymptomatic properties and high treatment resistance of ovarian cancer result in poor treatment outcomes and high mortality rates. Although the fundamental chemotherapy provides promising anticancer activities, it is associated with severe side effects. The derivative of phenothiazine, namely, 10H-3,6-diazaphenothiazine (PTZ), was synthesized and reported with ideal anticancer effects in a previous paper. In this study, detailed anticancer properties of PTZ was examined on A2780 ovarian cancer cells by investigating the cytotoxicity profiles, mechanism of apoptosis, and cell invasion. Research outcomes revealed PTZ-induced dose-dependent inhibition on A2780 cancer cells (IC50 =0.62 µM), with significant less cytotoxicity toward HEK293 normal kidney cells and H9C2 normal heart cells. Generation of reactive oxygen species (ROS) and polarization of mitochondrial membrane potential (ΔΨm) suggests PTZ-induced cell death through oxidative damage. The RT2 Profiler PCR Array on apoptosis pathway demonstrated PTZ-induced apoptosis via intrinsic (mitochondria-dependent) and extrinsic (cell death receptor-dependent) pathway. Inhibition of NF-κB and subsequent inhibition of (BIRC6-XIAP) complex activities reduced the invasion rate of A2780 cancer cells penetrating through the Matrigel™ Invasion Chamber. Lastly, the cell cycle analysis hypothesizes that the compound is cytostatic and significantly arrests cell proliferation at G2/M phase. Hence, the exploration of the underlying anticancer mechanism of PTZ suggested its usage as promising chemotherapeutic agent.
Collapse
Affiliation(s)
- Jianxin Zhang
- Department of Gynecology and Obstetrics, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing
| | - Chen Ming
- Department of Gynecologic Oncology, Taizhou People's Hospital, Jiangsu, People's Republic of China
| | | | | | - Beata Morak-Młodawska
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Krystian Pluta
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Małgorzata Jeleń
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Abdah Md Akim
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang
| | | | - Kah Kooi Ooi
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang.,Research Centre for Crystaline Materials, School of Science and Technology, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
41
|
Herzog TJ, Ison G, Alvarez RD, Balasubramaniam S, Armstrong DK, Beaver JA, Ellis A, Tang S, Ford P, McKee A, Gershenson DM, Kim G, Monk BJ, Pazdur R, Coleman RL. FDA ovarian cancer clinical trial endpoints workshop: A Society of Gynecologic Oncology White Paper. Gynecol Oncol 2017; 147:3-10. [PMID: 28844539 DOI: 10.1016/j.ygyno.2017.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/05/2017] [Accepted: 08/08/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Thomas J Herzog
- University of Cincinnati Cancer Institute, Dept. of Ob/Gyn, University of Cincinnati, United States
| | - Gwynn Ison
- Office of Hematology Oncology Products, OND, CDER, FDA, United States
| | - Ronald D Alvarez
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | | | - Julia A Beaver
- Office of Hematology Oncology Products, OND, CDER, FDA, United States
| | - Annie Ellis
- Ovarian Cancer Survivor, White Plains, New York
| | - Shenghui Tang
- Division of Biometrics V, OB, OTS, CDER, FDA, United States
| | - Peg Ford
- Ovarian Cancer Alliance of San Diego, United States
| | - Amy McKee
- Office of Hematology Oncology Products, OND, CDER, FDA, United States
| | | | - Geoffrey Kim
- Office of Hematology Oncology Products, OND, CDER, FDA, United States
| | | | | | - Robert L Coleman
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
42
|
Fields EC, McGuire WP, Lin L, Temkin SM. Radiation Treatment in Women with Ovarian Cancer: Past, Present, and Future. Front Oncol 2017; 7:177. [PMID: 28871275 PMCID: PMC5566993 DOI: 10.3389/fonc.2017.00177] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/02/2017] [Indexed: 01/31/2023] Open
Abstract
Ovarian cancer is the most lethal of the gynecologic cancers, with 5-year survival rates less than 50%. Most women present with advanced stage disease as the pattern of spread is typically with dissemination of malignancy throughout the peritoneal cavity prior to development of any symptoms. Prior to the advent of platinum-based chemotherapy, radiotherapy was used as adjuvant therapy to sterilize micrometastatic disease. The evolution of radiotherapy is detailed in this review, which establishes radiotherapy as an effective therapy for women with micrometastatic disease in the peritoneal cavity after surgery, ovarian clear cell carcinoma, focal metastatic disease, and for palliation of advanced disease. However, with older techniques, the toxicity of whole abdominal radiotherapy and the advancement of systemic therapies have limited the use of radiotherapy in this disease. With newer radiotherapy techniques, including intensity-modulated radiotherapy (IMRT), stereotactic body radiotherapy (SBRT), and low-dose hyperfractionation in combination with targeted agents, radiotherapy could be reconsidered as part of the standard management for this deadly disease.
Collapse
Affiliation(s)
- Emma C Fields
- Division of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - William P McGuire
- Internal Medicine, Virginia Commonwealth University, Richmond VA, United States
| | - Lilie Lin
- Division of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Sarah M Temkin
- Division of Gynecologic Oncology, Virginia Commonwealth University, Main Hospital, Richmond, VA, United States
| |
Collapse
|
43
|
Zhang H, Lu J, Lu Y, Zhou J, Wang Z, Liu H, Xu C. Prognostic significance and predictors of the system inflammation score in ovarian clear cell carcinoma. PLoS One 2017; 12:e0177520. [PMID: 28498842 PMCID: PMC5428928 DOI: 10.1371/journal.pone.0177520] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/29/2017] [Indexed: 01/04/2023] Open
Abstract
Chronic inflammation is a well-known epidemiologic factor of ovarian clear cell carcinomas (OCCC), but has an uncertain role in prognosis. We developed a systemic inflammation score (SIS) based on preoperative serum albumin and neutrophil-to-lymphocyte ratio (NLR) for predicting progression-free survival (PFS) and overall survival (OS) in OCCC patients. A retrospective review was performed in 155 patients with OCCC undergoing primary debulking and chemotherapy at a single institute between 1995 and 2010. Cox regression models were fitted to analyze the effect of prognostic factors on PFS and OS. Harrell’s concordance index was calculated to assess predictive accuracy. The SIS consisting of serum albumin and NLR was retained as an independent indicator adjusting for traditional clinicopathological features. A high SIS was significantly associated with aggressive tumor behavior, platinum resistance, and served as an independent predictor of reduced PFS (P = 0.006) and OS (P = 0.019). The SIS had a good discrimination ability for the predictive PFS (c-index = 0.712) and OS (c-index = 0.722). We have developed a system inflammation score for predicting prognosis of OCCC patients, which may help stratify patients for postsurgical management.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Gynaecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiaqi Lu
- Department of Gynaecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yingying Lu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jiayi Zhou
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Zehua Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- * E-mail: (HL); (CX)
| | - Congjian Xu
- Department of Gynaecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- * E-mail: (HL); (CX)
| |
Collapse
|
44
|
Baert T, Vergote I, Coosemans A. Ovarian cancer and the immune system. Gynecol Oncol Rep 2017; 19:57-58. [PMID: 28127584 PMCID: PMC5247278 DOI: 10.1016/j.gore.2017.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 01/16/2023] Open
Abstract
Short communication in response to the review of Turner et al. entitled “Ovarian cancer and the immune system - the role of targeted therapies” published in Gynecological Oncology. We believe systemic immune parameters might be a good alternative to tumor biopsy to gain insight in the immunological background of ovarian cancer. The immune system is an important player in ovarian cancer behaviour. Intratumoral studies of the immune system show an overwhelming immunosuppression. The immune signature in the blood can be important as a new biomarker.
Collapse
Affiliation(s)
- Thaïs Baert
- Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium; Department of Oncology, Laboratory for Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven, Belgium
| | - Ignace Vergote
- Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium; Department of Oncology, Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - An Coosemans
- Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium; Department of Oncology, Laboratory for Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Xiang J, Zhou L, Li X, Bao W, Chen T, Xi X, He Y, Wan X. Preoperative Monocyte-to-Lymphocyte Ratio in Peripheral Blood Predicts Stages, Metastasis, and Histological Grades in Patients with Ovarian Cancer. Transl Oncol 2016; 10:33-39. [PMID: 27888711 PMCID: PMC5124360 DOI: 10.1016/j.tranon.2016.10.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
PURPOSE: The monocyte-to-lymphocyte ratio (MLR) has been shown to be associated with the prognosis of various solid tumors. This study sought to evaluate the important value of the MLR in ovarian cancer patients. METHODS: A total of 133 ovarian cancer patients and 43 normal controls were retrospectively reviewed. The patients' demographics were analyzed along with clinical and pathologic data. The counts of peripheral neutrophils, lymphocytes, monocytes, and platelets were collected and used to calculate the MLR, neutrophil-to-lymphocyte ratio (NLR). and platelet-to-lymphocyte ratio (PLR). The optimal cutoff value of the MLR was determined by using receiver operating characteristic curve analysis. We compared the MLR, NLR, and PLR between ovarian cancer and normal control patients and among patients with different stages and different grades, as well as between patients with lymph node metastasis and non–lymph node metastasis. We then investigated the value of the MLR in predicting the stage, grade, and lymph node positivity by using logistic regression. The impact of the MLR on overall survival (OS) was calculated by Kaplan-Meier method and compared by log-rank test. RESULTS: Statistically significant differences in the MLR were observed between ovarian cancer patients and normal controls. However, no difference was found for the NLR and PLR. Highly significant differences in the MLR were found among patients with different stages (stage I-II and stage III-IV), grades (G1 and >G1), and lymph node metastasis status. The MLR was a significant and independent risk factor for lymph node metastasis, as determined by logistic regression. The optimal cutoff value of the MLR was 0.23. We also classified the data according to tumor markers (CA125, CA199, HE4, AFP, and CEA) and conventional coagulation parameters (International Normalized Ratio [INR] and fibrinogen). Highly significant differences in CA125, CA199, HE4, INR, fibrinogen levels, and lactate dehydrogenase were found between the low-MLR group (MLR ≤ 0.23) and the high-MLR group (MLR > 0.23). Correspondingly, dramatic differences were observed between the two groups in OS. CONCLUSION: Our results show that the peripheral blood MLR before surgery could be a significant predictor of advanced stages, advanced pathologic grades, and positive lymphatic metastasis in ovarian cancer patients.
Collapse
Affiliation(s)
- Jiangdong Xiang
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Lina Zhou
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xing Li
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Wei Bao
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Taizhong Chen
- F1370016, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xiaowei Xi
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| | - Yinyan He
- Department of Obstetrics and Gynaecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| | - Xiaoping Wan
- Shanghai First Maternity and Infant Hospital Corporation, Tongji University, Shanghai, 200126, China
| |
Collapse
|