1
|
Praiss AM, Moukarzel LA, Zhu Y, Longhini ALF, Derakhshan F, Hoang T, Pesci G, Green H, Ozsoy MA, Hanlon E, Kahn R, Brodeur MN, Sia T, Abu-Rustum NR, Gardner G, Roche KL, Sonoda Y, Zivanovic O, Chi DS, Merghoub T, Gardner R, Weigelt B, Zamarin D. Evolution of tumor stress response during cytoreductive surgery for ovarian cancer. iScience 2025; 28:112317. [PMID: 40256326 PMCID: PMC12008711 DOI: 10.1016/j.isci.2025.112317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Upfront treatment for patients with advanced high-grade serous ovarian cancer (HGSOC) includes a multi-hour cytoreductive surgery. Although the procedure is necessary for maximal tumor cytoreduction, understanding of the biology of systemic and intratumoral responses induced by surgical cytoreduction is limited. Through analysis of matched tumor and normal tissues and peripheral blood collected at multiple time points during cytoreductive surgery in patients with HGSOC, we demonstrate that surgery leads to rapid induction of systemic inflammatory response and activation of inflammatory signaling in the tumor and normal tissue, with interleukin-6 emerging as a dominant inflammatory pathway. A parallel study in a syngeneic murine HGSOC model recapitulated these findings and demonstrated accelerated tumor growth in response to surgery. This study highlights the previously unappreciated impact of specimen collection timing on the tumor signaling networks and provides insights into stress pathways activated by surgery, generating rationale for perioperative therapeutic interventions to reduce protumorigenic effects.
Collapse
Affiliation(s)
- Aaron M. Praiss
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lea A. Moukarzel
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingjie Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Leda F. Longhini
- Department of Flow Cytometry, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fatemeh Derakhshan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy Hoang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giulio Pesci
- Ludwig Collaborative Laboratory, Weill Cornell Medicine, New York, NY, USA
| | - Hunter Green
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melih A. Ozsoy
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Etta Hanlon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan Kahn
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Tiffany Sia
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem R. Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Ginger Gardner
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Kara Long Roche
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Zivanovic
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis S. Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative Laboratory, Weill Cornell Medicine, New York, NY, USA
| | - Rui Gardner
- Department of Flow Cytometry, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Roberts AL, Townsend MK, Chibnik LB, Kubzansky LD, Tworoger SS. Timing of depression in relation to risk of ovarian cancer. J Natl Cancer Inst 2025; 117:989-996. [PMID: 39745894 PMCID: PMC12058258 DOI: 10.1093/jnci/djae348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/19/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Several studies have suggested that depression may be associated with increased risk of ovarian cancer. Less is known about whether timing matters regarding when depression occurs. To provide evidence for an etiologically relevant exposure period, we examined depression occurring during the time in which precursor lesions develop and progress to invasive carcinoma with the risk of developing ovarian cancer. METHODS Using data from 2 prospective cohorts (1992-2015), we divided follow-up into consecutive 2-year periods for analytic purposes, referred to as "cancer ascertainment periods." We estimated associations of depression in the 10 years before each cancer ascertainment period with incident cancer, using Cox proportional hazards models. Next, we estimated associations of depression occurring up to 18 years before each ascertainment period, in 2-year increments, with incident cancer. We adjusted for demographic, health, and behavioral factors. All tests of statistical significance were two-sided, with a P-value threshold of less than .05. RESULTS Depression occurring in the 10 prior years was associated with 30% greater risk of cancer (hazard ratio [HR] = 1.30, 95% confidence interval = 1.15 to 1.46). Associations were similar in fully adjusted models (HR = 1.27). Depression occurring in the 14 years before ascertainment was associated with elevated risk, although only estimates for depression 0-2, 6-8, and 8-10 years before ascertainment reached statistical significance (HR range = 1.20-1.36). CONCLUSION Depression occurring up to 14 years before cancer ascertainment was associated with greater cancer risk. This is the time of precursor progression to invasive ovarian carcinoma, suggesting depression may be an ovarian cancer-promoting agent.
Collapse
Affiliation(s)
- Andrea L Roberts
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Mary K Townsend
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, United States
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Lori B Chibnik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Shelley S Tworoger
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, United States
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
3
|
Ma Z, Zhang J, Li Z, Zhu Y, Han X, Lei L, Cheng K, Liu W. Interleukin-1β Inhibits Ovarian Cancer Cell Proliferation and Metastasis Through the MAPK/MMP12 Pathway. Int J Mol Sci 2025; 26:3287. [PMID: 40244135 PMCID: PMC11989259 DOI: 10.3390/ijms26073287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecological tumor with high mortality. Despite aggressive treatment, survival rates for patients with advanced EOC are low, and more effective methods of diagnosis and treatment are urgently needed. Inflammation and cancer are strongly associated; however, the mechanisms that mediate this relationship are not fully understood. In this study, we found that the expression of interleukin-1β (IL-1β), a proinflammatory cytokine, increased in an ovarian cancer tissue microarray (TMA) and inhibited A2780 and SKOV3 cell viability and metastasis. Recombinant IL-1β protein and the overexpression of IL-1β decreased the proliferation and metastasis of ovarian cancer cells. IL-1β deficiency promoted proliferation and metastasis. Moreover, transcriptome sequencing revealed that IL-1β downregulates the expression of matrix metalloproteinase 12 (MMP12). The signaling pathway involving MAPK/AP-1/MMP12 is involved in IL-1β-regulated ovarian cancer progression. Overall, we found that the proinflammatory cytokine IL-1β inhibits ovarian cancer cell viability and metastasis. These findings provided deeper insights into inflammation and cancer progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China; (Z.M.); (J.Z.); (Z.L.); (Y.Z.); (X.H.); (L.L.); (K.C.)
| |
Collapse
|
4
|
Tang Y, Zhang YY, Wen MB, Li L, Hu HQ, Zeng YH, Shi Q. Patient-reported symptom burden and circulating cytokines undergoing chemotherapy: a pilot study in patients with ovarian cancer. J Gynecol Oncol 2025; 36:e17. [PMID: 38991946 PMCID: PMC11964978 DOI: 10.3802/jgo.2025.36.e17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVE To analyze the fluctuations of patient-reported outcomes (PROs) and their relationships with cytokines in the peripheral blood of patients undergoing chemotherapy for ovarian cancer (OC). METHODS PROs burden was prospectively measured by the M.D. Anderson Symptom Inventory-Ovarian Cancer (MDASI-OC) at baseline before chemotherapy, on a daily basis during and post-chemotherapy days (PCD) 7, 14, and 20. Cytokines were collected at baseline, days prior to hospital discharge and PCD 20. Pearson correlation was used to explore the associations between PROs and cytokines levels in peripheral blood. RESULTS The top 8 rated symptoms were compared between the neoadjuvant chemotherapy (NACT) group (n=20) and the postoperative adjuvant chemotherapy (PAC) group (n=7). Before chemotherapy, the mean scores of fatigue and lack of appetite in the NACT group were higher than those in the PAC group. After chemotherapy, pain, nausea, vomiting, disturbed sleep, lack of appetite, and constipation increased to peak during PCD 2-6; while, fatigue and numbness or tingling remained at high levels over PCD 2-13. By PCD 20, disturbed sleep and fatigue showed a significant increase in mean scores, particularly in the NACT group; while, other symptom scores decreased and returned to baseline levels. Additionally, the longitudinal fluctuations in pain, fatigue, and lack of appetite were positively associated with circulating levels of interleukin-6 and interferon gamma (p<0.05). CONCLUSION MDASI-OC was feasible and adaptable for demonstrating the fluctuations of symptom burden throughout chemotherapy course. Moreover, symptoms changing along with cytokines levels could provide clues for exploring mechanism underlying biochemical etiology.
Collapse
Affiliation(s)
- Ying Tang
- Department of Obstetrics and Gynecology, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yuan-Yuan Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ming-Bo Wen
- Department of Obstetrics and Gynecology, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Lin Li
- Department of Clinical Laboratory, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Hui-Quan Hu
- Department of Obstetrics and Gynecology, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Yu-Hua Zeng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qiuling Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Wang J, Zhu W, Li X, Wu Y, Ma W, Wang Y, Zhao W, Wei F, Wang W. Transcriptome analysis of ovarian cancer uncovers association between tumor-related inflammation/immunity and patient outcome. Front Pharmacol 2025; 16:1500251. [PMID: 39981173 PMCID: PMC11839622 DOI: 10.3389/fphar.2025.1500251] [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: 09/23/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Background Epithelial ovarian cancer (EOC) is a cancer that affects the female reproductive system and is highly lethal. It poses significant challenges in terms of treatment and often has a poor prognosis. In recent years, with the advent of PARPi, the treatment of ovarian cancer has entered a new stage of full-process management. Although more and more drugs have been approved, the therapeutic effect of PARPi is still very limited. With the rapid development of PD-1/PD-L1, CTLA-4, oncolytic viruses, cancer vaccines, adoptive cell therapy, etc., tumor immunotherapy has provided new opportunities for the treatment of ovarian cancer. Methods This study used comprehensive transcriptome analysis across multiple databases to gather gene transcripts and clinical features of normal ovarian samples and tissue samples from ovarian cancer. The aim was to explore the mechanisms underlying tumor immunotherapy resistance and to reveal the relationship between ovarian cancer's immune microenvironment and genes linked to inflammation. Various R packages were used for differential gene analysis, enrichment analysis, co-expression network construction, and prognostic model building. Results It has been found that the prognosis of ovarian cancer patients is closely associated with sets of genes involved in inflammation. The immune infiltration microenvironment, clinicopathological features, and survival rates differed significantly between two inflammatory gene expression patterns identified using cluster and immune microenvironment analyses. Further analysis revealed that the high-risk group had a higher abundance of M2-type macrophage infiltration, more active anti-tumor immune response, higher tumor stemness score, potentially worse prognosis, and lower response rates to multiple chemotherapy drugs and immune checkpoint inhibitors. Conclusion These findings provide new perspectives and potential targets for immunotherapy and prognostic evaluation of ovarian cancer and offer new strategies and directions for clinical treatment and patient management. This study provides crucial information to further our comprehension of drug response mechanisms and tumor immunotherapy. It offers new strategies and methods for the treatment and prognostic improvement of ovarian cancer.
Collapse
Affiliation(s)
- Jingfang Wang
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenrui Zhu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xia Li
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuanyuan Wu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenhui Ma
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yangzhou Wang
- Department of Stomatology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Weihong Zhao
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fang Wei
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenhao Wang
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Wang YX, Farland LV, Gaskins AJ, Wang S, Terry KL, Rexrode KM, Rich-Edwards JW, Tamimi R, Chavarro JE, Missmer SA. Endometriosis and uterine fibroids and risk of premature mortality: prospective cohort study. BMJ 2024; 387:e078797. [PMID: 39567014 PMCID: PMC11577545 DOI: 10.1136/bmj-2023-078797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE To prospectively assess the effect of endometriosis and uterine fibroids on the long term risk of premature mortality (younger than 70 years). DESIGN Prospective cohort study SETTING: The Nurses' Health Study II, United States (1989-2019). PARTICIPANTS 110 091 women aged 25-42 years in 1989 without a history of hysterectomy before endometriosis or fibroids diagnosis, cardiovascular diseases, or cancer. MAIN OUTCOME MEASURES Hazard ratios (estimated by Cox proportional hazards models) for total and cause specific premature mortality according to laparoscopically confirmed endometriosis or ultrasound or hysterectomy confirmed uterine fibroids reported in biennial questionnaires. RESULTS 4356 premature deaths were recorded during 2 994 354 person years of follow-up (27.2 years per person), including 1459 from cancer, 304 from cardiovascular diseases, and 90 from respiratory diseases. The crude incidence of all cause premature mortality for women with and without laparoscopically confirmed endometriosis was 2.01 and 1.40 per 1000 person years, respectively. In age adjusted models, laparoscopically confirmed endometriosis was associated with a hazard ratio of 1.19 (95% confidence interval 1.09 to 1.30) for premature death; these models were strengthened after also adjusting for potential confounders including behavioral factors (1.31, 1.20 to 1.44). Cause specific mortality analyses showed that the association was largely driven by mortality from senility and ill-defined diseases (1.80, 1.19 to 2.73), non-malignant respiratory diseases (1.95, 1.11 to 3.41), diseases of the nervous system and sense organs (2.50, 1.40 to 4.44), and malignant neoplasm of gynecological organs (2.76, 1.79 to 4.26). Ultrasound or hysterectomy confirmed uterine fibroids were not associated with all cause premature mortality (1.03, 0.95 to 1.11), but were associated with a greater risk of mortality from malignant neoplasm of gynecological organs (2.32, 1.59 to 3.40) in cause specific mortality analyses. The risk of mortality caused by cardiovascular and respiratory diseases varied according to joint categories of endometriosis and uterine fibroids, with an increased risk of all cause premature mortality among women reporting both endometriosis and uterine fibroids. CONCLUSION Women with a history of endometriosis and uterine fibroids might have an increased long term risk of premature mortality extending beyond their reproductive lifespan. These conditions were also associated with an increased risk of death due to gynecological cancers. Endometriosis was associated with a greater risk of non-cancer mortality. These findings highlight the importance for primary care providers to consider these gynecological disorders in their assessment of women's health.
Collapse
Affiliation(s)
- Yi-Xin Wang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health and Department of Obstetrics and Gynecology, College of Medicine-Tucson, University of Arizona, Tucson, AZ, USA
| | - Audrey J Gaskins
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Siwen Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L Terry
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Janet W Rich-Edwards
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rulla Tamimi
- Division of Epidemiology, Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stacey A Missmer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics and Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
7
|
Fu YP, Lin H, Ou YC, Wu CH, Fu HC. Bevacizumab as a mitigating factor for the impact of high systemic immune-inflammation index on chemorefractory in advanced epithelial ovarian cancer. BMC Cancer 2024; 24:1377. [PMID: 39529011 PMCID: PMC11552161 DOI: 10.1186/s12885-024-13087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Predicting chemorefractory disease in advanced epithelial ovarian cancer (EOC) remains challenging. This study aimed to identify clinicopathological factors and hemogram data as predictive markers for chemorefractory EOC and to explore potential therapeutic approaches that may mitigate these unfavorable conditions. METHODS We conducted a retrospective analysis of patients with advanced EOC treated with chemotherapy. Hemogram data and clinicopathological variables were collected. We employed logistic regression to assess factors associated with chemorefractory EOC and used the Kaplan-Meier method for survival analysis. RESULTS Among the 191 patients analyzed, suboptimal surgery, lymphocyte count < 1440/mm3, systemic immune-inflammation index (SII) ≥ 2350, and lack of bevacizumab therapy were independently associated with chemorefractory EOC (OR 19.30, 95% CI 7.01-53.12; OR 9.07, 95% CI 2.76-29.82; OR 12.45, 95% CI 3.87-40.07; OR 6.61, 95% CI 2.01-21.78, respectively). Elevated SII was also identified as a risk factor for poor progression-free (PFS) and overall survival (OS). Specifically, patients with high SII who did not receive bevacizumab had a significantly higher probability of chemorefractory EOC and poorer survival outcomes compared to those who received bevacizumab. CONCLUSIONS Our findings suggest that hemogram parameters and clinicopathological factors such as suboptimal surgery, lymphocyte count, SII, and bevacizumab therapy status are predictive markers for chemorefractory disease in advanced EOC. Elevated SII emerged as a predictor for poorer PFS and OS outcomes, particularly in the absence of bevacizumab therapy.
Collapse
Affiliation(s)
- Yan-Ping Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
8
|
Zhang M, Mo J, Huang W, Bao Y, Luo X, Yuan L. The ovarian cancer-associated microbiome contributes to the tumor's inflammatory microenvironment. Front Cell Infect Microbiol 2024; 14:1440742. [PMID: 39497925 PMCID: PMC11532186 DOI: 10.3389/fcimb.2024.1440742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
A growing body of research has established a correlation between tumors and persistent chronic inflammatory infiltration. As a primary instigator of inflammation, the majority of microbiomes naturally residing within our bodies engage in a mutually beneficial symbiotic relationship. Nevertheless, alterations in the microbiome's composition or breaches in the normal barrier function can disrupt the internal environment's homeostasis, potentially leading to the development and progression of various diseases, including tumors. The investigation of tumor-related microbiomes has contributed to a deeper understanding of their role in tumorigenesis. This review offers a comprehensive overview of the microbiome alterations and the associated inflammatory changes in ovarian cancer. It may aid in advancing research to elucidate the mechanisms underlying the ovarian cancer-associated microbiome, providing potential theoretical support for the future development of microbiome-targeted antitumor therapies and early screening through convenient methods.
Collapse
Affiliation(s)
- Min Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiahang Mo
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wu Huang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yiting Bao
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xukai Luo
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Yuan
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Lazar A, Popa AM, Orlov-Slavu C, Cotan HT, Iaciu CI, Olaru CM, Schreiner OD, Ciobanu RC, Nitipir C. The Influence of Circulating Immune Cell and CA125 Dynamics on Neoadjuvant Therapy Selection for Advanced Ovarian Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1290. [PMID: 39202571 PMCID: PMC11356539 DOI: 10.3390/medicina60081290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Ovarian cancer, including tubal and peritoneal cancer, is the third most common gynecological cancer and the leading cause of death from gynecological malignancies in developed countries. This study explores the prognostic value of the neutrophil-to-lymphocyte ratio (NLR) in determining the optimal duration of neoadjuvant chemotherapy (NACT) for advanced ovarian cancer. It also investigates the correlation between NLR dynamics and the KELIM score, a chemosensitivity marker, to enhance individualized therapeutic strategies. Materials and Methods: A total of 79 patients underwent NACT followed by interval debulking surgery (IDS) or palliative care. The data collected included demographic information, tumor characteristics, treatment modalities, and laboratory parameters. The baseline NLR (NLR-T0) and post-therapeutic NLR (NLR-T1) were calculated, and their variation (NLR∆) was analyzed. The KELIM score was determined using CA-125 values. Results: Patients with a high baseline NLR (≥2.5) had significantly worse progression-free survival (PFS) and overall survival (OS) compared to those with a low NLR (<2.5). A negative NLR∆ was associated with poorer PFS and OS. The KELIM score indicated a more effective treatment response, with higher scores correlating with better outcomes. The majority of patients achieved R0 resection, and those with favorable KELIM scores showed improved survival rates. Conclusions: The NLR is a valuable prognostic marker for assessing treatment response and guiding NACT duration in advanced ovarian cancer.
Collapse
Affiliation(s)
- Alexandra Lazar
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Ana Maria Popa
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Cristina Orlov-Slavu
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Horia-Teodor Cotan
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Cristian Ion Iaciu
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Cristina Mihaela Olaru
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| | - Oliver Daniel Schreiner
- Regional Institute of Oncology Iasi, 2-4 General Henri Mathias Berthelot Street, 700483 Iasi, Romania;
- Department 3—Medical Sciences, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Gheorghe Asachi Technical University, 700050 Iasi, Romania
| | - Romeo Cristian Ciobanu
- Department of Electrical Measurements and Materials, Gheorghe Asachi Technical University, 700050 Iasi, Romania
| | - Cornelia Nitipir
- Department 8—Clinical Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (A.L.); (A.M.P.); (C.O.-S.); (H.-T.C.); (C.I.I.); (C.M.O.); (C.N.)
- Department of Medical Oncology, Elias Emergency University Hospital, 17 Bd. Marasti, 011461 Bucharest, Romania
| |
Collapse
|
10
|
Li H, Li B, Wang X, Zhang H, Wang C, Fan B, Wang L. Effect of longitudinal changes of cachexia on the efficacy and toxicity of immune checkpoint inhibitors in esophageal squamous cell cancer (ESCC) patients. Nutrition 2024; 124:112462. [PMID: 38663128 DOI: 10.1016/j.nut.2024.112462] [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: 01/08/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 06/17/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have enhanced survival in advanced esophageal squamous cell cancer (ESCC) patients, but their efficacy varies. Cachexia, characterized by muscle loss and significant weight loss, might influence ICI response. This study examines the relationship between cachexia's longitudinal changes and ICI outcomes in ESCC patients. METHODS ESCC patients undergoing at least two ICI cycles from 2017 to 2021 were studied. Cachexia's baseline and evolving patterns during ICI treatment were observed. Kaplan-Meier and Cox regression analyses were used to assess cachexia's effect on ICI efficacy. Chi-square tests were used to determine cachexia's link to immune-related adverse effects (irAEs). RESULTS Two hundred seventy-eight ICI-treated patients had a median progression-free survival (PFS) of 5.78 months and overall survival (OS) of 8.3 months. Pretreatment cachexia led to worse outcomes: PFS 7.87 versus 5.3 months, time to progression (TTP) 10.9 versus 6.1 months, and OS 14.3 versus 9.2 months. Irreversible cachexia showed the poorest results. Cachexia's changes weren't associated with irAEs. CONCLUSION Baseline and evolving cachexia significantly impact ICI efficacy in ESCC patients. Continuous cachexia monitoring during ICI therapy is crucial for optimal ESCC management.
Collapse
Affiliation(s)
- Haoqian Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Butuo Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoqing Wang
- Department of Portal Hypertension, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Huan Zhang
- Department of Nursing, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chunni Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bingjie Fan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
11
|
Boidin L, Moinard M, Moussaron A, Merlier M, Moralès O, Grolez GP, Baydoun M, Mohd-Gazzali A, Tazizi MHDM, Allah HHA, Kerbage Y, Arnoux P, Acherar S, Frochot C, Delhem N. Targeted Photodynamic Therapy using a Vectorized Photosensitizer coupled to Folic Acid Analog induces Ovarian Tumor Cell Death and inhibits IL-6-mediated Inflammation. J Control Release 2024; 371:351-370. [PMID: 38789088 DOI: 10.1016/j.jconrel.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/10/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Ovarian cancer (OC) is one of the most lethal cancers among women. Frequent recurrence in the peritoneum due to the presence of microscopic tumor residues justifies the development of new therapies. Indeed, our main objective is to develop a targeted photodynamic therapy (PDT) treatment of peritoneal carcinomatosis from OC to improve the life expectancy of cancer patients. Herein, we propose a targeted-PDT using a vectorized photosensitizer (PS) coupled with a newly folic acid analog (FAA), named PSFAA, in order to target folate receptor alpha (FRα) overexpressed on peritoneal metastasis. This PSFAA was the result of the coupling of pyropheophorbide-a (Pyro-a), as the PS, to a newly synthesized FAA via a polyethylene glycol (PEG) spacer. The selectivity and the PDT efficacy of PSFAA was evaluated on two human OC cell lines overexpressing FRα compared to fibrosarcoma cells underexpressing FRα. Final PSFAA, including the synthesis of a newly FAA and its conjugation to Pyro-a, was obtained after 10 synthesis steps, with an overall yield of 19%. Photophysical properties of PSFAA in EtOH were performed and showed similarity with those of free Pyro-a, such as the fluorescence and singlet oxygen quantum yields (Φf = 0.39 and ΦΔ = 0.53 for free Pyro-a, and Φf = 0.26 and ΦΔ = 0.41 for PSFAA). Any toxicity of PSFAA was noticed. After light illumination, a dose-dependent effect on PS concentration and light dose was shown. Furthermore, a PDT efficacy of PSFAA on OC cell secretome was detected inducing a decrease of a pro-inflammatory cytokine secretion (IL-6). This new PSFAA has shown promising biological properties highlighting the selectivity of the therapy opening new perspectives in the treatment of a cancer in a therapeutic impasse.
Collapse
Affiliation(s)
- Léa Boidin
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Morgane Moinard
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France
| | - Albert Moussaron
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France; Univ. Lorraine, CNRS, UMR7375 - LCPM - Laboratoire de Chimie-Physique Macromoléculaire, Nancy F-54000, France
| | - Margaux Merlier
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Olivier Moralès
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France; Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France
| | - Guillaume Paul Grolez
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Martha Baydoun
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Amirah Mohd-Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Hassan Hadi Abd Allah
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Yohan Kerbage
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Philippe Arnoux
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France
| | - Samir Acherar
- Univ. Lorraine, CNRS, UMR7375 - LCPM - Laboratoire de Chimie-Physique Macromoléculaire, Nancy F-54000, France.
| | - Céline Frochot
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France.
| | - Nadira Delhem
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France.
| |
Collapse
|
12
|
Zhang Z, Lang J. The prognostic and clinical value of neutrophil-to-lymphocyte ratio (NLR) in ovarian cancer: A systematic review and meta-analysis. J Med Biochem 2024; 43:323-333. [PMID: 39139167 PMCID: PMC11318050 DOI: 10.5937/jomb0-46035] [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: 07/14/2023] [Accepted: 10/17/2023] [Indexed: 08/15/2024] Open
Abstract
Background Ovarian cancer (OC) is a major gynecological malignancy with varying prognosis. The Neutrophil-toLymphocyte Ratio (NLR) has been proposed as a potential prognostic biomarker. This study aimed to evaluate the prognostic and clinical value of NLR in OC. Methods A systematic review and meta-analysis were performed following PRISMA guidelines, including studies that evaluated the association between NLR and survival outcomes in OC patients. Search was performed in PubMed, Embase, Web of Science, and Cochrane Library databases. Quality assessment was done using Newcastle-Ottawa Scale (NOS). Heterogeneity was assessed, and pooled hazard ratios (HRs) were calculated using fixed or random-effects models as appropriate.
Collapse
Affiliation(s)
- Zihan Zhang
- Chinese Academy of Medical Sciences, Union Medical College, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Jinghe Lang
- Chinese Academy of Medical Sciences, Union Medical College, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
13
|
Calmon MS, Lemos FFB, Silva Luz M, Rocha Pinheiro SL, de Oliveira Silva LG, Correa Santos GL, Rocha GR, Freire de Melo F. Immune pathway through endometriosis to ovarian cancer. World J Clin Oncol 2024; 15:496-522. [PMID: 38689629 PMCID: PMC11056862 DOI: 10.5306/wjco.v15.i4.496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/22/2024] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disease, defined by the presence of functional endometrial tissue outside of the uterine cavity. This disease is one of the main gynecological diseases, affecting around 10%-15% women and girls of reproductive age, being a common gynecologic disorder. Although endometriosis is a benign disease, it shares several characteristics with invasive cancer. Studies support that it has been linked with an increased chance of developing endometrial ovarian cancer, representing an earlier stage of neoplastic processes. This is particularly true for women with clear cell carcinoma, low-grade serous carcinoma and endometrioid. However, the carcinogenic pathways between both pathologies remain poorly understood. Current studies suggest a connection between endometriosis and endometriosis-associated ovarian cancers (EAOCs) via pathways associated with oxidative stress, inflammation, and hyperestrogenism. This article aims to review current data on the molecular events linked to the development of EAOCs from endometriosis, specifically focusing on the complex relationship between the immune response to endometriosis and cancer, including the molecular mechanisms and their ramifications. Examining recent developments in immunotherapy and their potential to boost the effectiveness of future treatments.
Collapse
Affiliation(s)
- Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Reis Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
14
|
Tuntinarawat P, Tangmanomana R, Kittisiam T. Association between alteration of neutrophil to lymphocyte ratio, platelet to lymphocyte ratio, cancer antigen-125 and surgical outcomes in advanced stage ovarian cancer patient who received neoadjuvant chemotherapy. Gynecol Oncol Rep 2024; 52:101347. [PMID: 38419812 PMCID: PMC10899061 DOI: 10.1016/j.gore.2024.101347] [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: 01/10/2024] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Optimal resection significantly influences the prognosis of advanced-stage epithelial ovarian cancer (EOC) patients undergoing debulking surgery. In patients who received neoadjuvant chemotherapy (NACT) followed by interval debulking surgery (IDS), the determination of the ideal timing for surgery remains a challenge. Inflammatory markers, including the neutrophil-to-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and CA-125 levels, have been recognized as potential predictive markers. Objective This study aims to evaluate the predictive value of changes in NLR, PLR, and CA-125 levels following NACT, specifically assessing their impact on surgical outcomes during IDS for advanced-stage EOC. Methods A retrospective cohort study enrolled advanced-stage EOC patients who underwent NACT followed by IDS at Vajira Hospital in Thailand from January 2009 to June 2023. Data on clinical, surgical, and inflammatory markers were collected, and the predictive value of these markers for suboptimal resection outcomes was assessed. Results Among the 65 patients, 98.5 % exhibited radiologic responses post-NACT, while 29.2 % experienced suboptimal resections. Univariate analysis did not reveal significant associations between suboptimal resection and NLR changes after the first NACT cycle or alterations in NLR, PLR, and CA-125 levels at the end of NACT. Subsequent analysis suggested that an NLR decrease exceeding 70 % after the first cycle and NACT completion might predict suboptimal resection, yet statistical analyses showed limited prognostic efficacy (AuROC = 0.608 and 0.597). Conclusion Our study does not support that changes in NLR, PLR, platelet count, and CA-125 levels after NACT reliably predict IDS outcomes. Additional prospective investigations using larger cohorts or a combination of evaluation methods, rather than relying solely on NLR, are recommended.
Collapse
Affiliation(s)
- Ponganun Tuntinarawat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Ratnapat Tangmanomana
- Department of Obstetrics and Gynecology, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Thannaporn Kittisiam
- Department of Obstetrics and Gynecology, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| |
Collapse
|
15
|
Krasny S, Baranau Y, Polyakov S, Zharkova E, Streltsova O, Filimonava A, Siarheyeva V, Kazlouskaya S, Khorau A, Gabai V, Shneider A. Clinical efficacy of plasmid encoding p62/SQSTM1 (Elenagen) in combination with gemcitabine in patients with platinum-resistant ovarian cancer: a randomized controlled trial. Front Oncol 2024; 14:1343023. [PMID: 38410116 PMCID: PMC10895999 DOI: 10.3389/fonc.2024.1343023] [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: 11/22/2023] [Accepted: 01/12/2024] [Indexed: 02/28/2024] Open
Abstract
Background The purpose of this trial is to evaluate the safety and efficacy of ELENAGEN, a novel anticancer therapeutic DNA plasmid encoding p62/SQSTM1 protein, as an adjuvant to chemotherapy with gemcitabine (GEM) in patients with advanced platinum-resistant ovarian cancer. Methods This open-label prospective randomized study with two arms. GEM (1000 mg/m2) on days 1 and 8 every 3 weeks was administered in both arms: in the Chemo arm (n = 20), GEM was the only treatment, and in the ELENAGEN arm (n = 20), GEM was supplemented with ELENAGEN (2.5 mg i.m. weekly). The primary endpoint was progression-free survival (PFS), and the secondary endpoint was safety. Antitumor activity was assessed by RECIST 1.1, and criteria safety was assessed according to NCI CTCAE version 5.0. Results According to the cutoff data, the median follow-up was 13.8 months. There were no serious adverse events related to ELENAGEN treatment. The median PFS was 2.8 and 7.2 months in the Chemo and ELENAGEN arms, respectively (p Log-Rank = 0.03). Notably, at the time of cutoff, 9 patients (45%) in the ELENAGEN arm did not progress, with the longest PFS recorded thus far being 24 months. Subgroup analysis of patients in both arms demonstrated high efficacy of ELENAGEN in patients with worse prognostic factors: high pretreatment levels of CA125 and progression after platinum-free interval <3 months. Conclusions The addition of ELENAGEN to gemcitabine is effective in patients with platinum-resistant ovarian cancer, including those with a worse prognosis. Clinical trial registration https://www.clinicaltrials.gov/study/NCT05979298, identifier NCT05979298, 2023-08-07.
Collapse
Affiliation(s)
- Sergei Krasny
- N. N. Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | | | - Sergey Polyakov
- N. N. Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | | | | | | | - Volha Siarheyeva
- N. N. Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | | | - Anton Khorau
- N. N. Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | | | - Alexander Shneider
- CureLab Oncology, Inc., Boston, MA, United States
- Department of Molecular Biology, Ariel University, Ariel, Israel
| |
Collapse
|
16
|
Zhu J, Han M, Yang Y, Feng R, Hu Y, Wang Y. Exploring the Mechanism of Brucea Javanica against Ovarian Cancer based on Network Pharmacology and the Influence of Luteolin on the PI3K/AKT Pathway. Comb Chem High Throughput Screen 2024; 27:157-167. [PMID: 37366364 DOI: 10.2174/1386207326666230627114111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Ovarian cancer (OC) is a commonly diagnosed female cancer around the world. The Chinese herbal medicine Brucea Javanica has an anti-cancer effect. However, there is no relevant report on whether Brucea Javanica is effective in treating OC, and the corresponding mechanism is also unknown. OBJECTIVE This study was projected to excavate the active components and underpinned molecular mechanisms of Brucea Javanica in treating ovarian cancer (OC) through network pharmacology combined with in vitro experiments. METHODS The essential active components of Brucea Javanica were selected using the TCMSP database. The OC-related targets were selected by GeneCards, intersecting targets were obtained by Venn Diagram. The core targets were obtained through the PPI network and Cytoscape, and the key pathway was gained through GO and KEGG enrichment analyses. Meanwhile, docking conformation was observed as reflected by molecular docking. MTT, colony formation assay and flow cytometer (FCM) analysis were performed to determine cell proliferation and apoptosis, respectively. Finally, Levels of various signaling proteins were evaluated by western blotting. RESULTS Luteolin, β-sitosterol and their corresponding targets were selected as the essential active components of Brucea Javanica. 76 intersecting targets were obtained by Venn Diagram. TP53, AKT1, and TNF were obtained through the PPI network and Cytoscape, and the key pathway PI3K/AKT was gained through GO and KEGG enrichment analyses. A good docking conformation was observed between luteolin and AKT1. Luteolin could hinder A2780 cell proliferation, induce cell apoptosis and enhance the inhibition of the PI3K/AKT pathway. CONCLUSION It was verified in vitro that luteolin could hinder OC cell proliferation and activate the PI3K/AKT pathway to lead to apoptosis.
Collapse
Affiliation(s)
- Jufan Zhu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Mengfei Han
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yiheng Yang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Renqian Feng
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yan Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yuli Wang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| |
Collapse
|
17
|
Schatten H. The Impact of Mitochondria in Ovarian Cancer Cell Metabolism, Proliferation, and Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1452:119-125. [PMID: 38805128 DOI: 10.1007/978-3-031-58311-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mitochondrial dysfunctions are significantly implicated in cancer initiation, progression, and metastasis, which have been shown for several cancers including ovarian cancer.An increase in mitochondrial dysfunction is also associated with drug resistance along with cancer progression, which in part is related to its specific microenvironment that is characterized by ascites, low glucose levels, and hypoxia that causes ovarian cancer cells to switch to mitochondrial respiration to enable their survival. Peritoneal ascitic fluid accumulation is a specific feature of ovarian cancer, and it is a major cause of its metastatic spread that also presents challenges for effective treatment. Among the treatment difficulties for ovarian cancer is the mutation rate and frequency of mtDNA in ovarian cancer tissue that can affect the efficiency of chemotherapeutic drugs. The varied and multiple mutations of different types enable metabolic reprogramming, cancer cell proliferation, and drug resistance.New specific information on mechanisms underlying several of the mitochondrial dysfunctions has led to proposing various mitochondrial determinants as targets for ovarian cancer therapy, which include targeting specific mitochondrial proteins and phosphoproteins as well as reactive oxygen species (ROS) that accumulate abnormally in cancer cells. Because of the genetically and histologically heterogeneous nature of the disease, combination therapy approaches will be necessary to combat the disease and achieve progress in effective treatment of ovarian cancer. This chapter will address (1) mitochondrial vulnerabilities underlying dysfunction and disease; (2) mitochondrial dysfunction in ovarian cancer; (3) present treatment difficulties for ovarian cancer and new potential treatment strategies to target ovarian cancer mitochondrial metabolism; and (4) biobehavioral factors influencing ovarian cancer development.
Collapse
Affiliation(s)
- Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri-Columbia, Columbia, MO, USA.
| |
Collapse
|
18
|
Santiago AE, Paula SOCD, Carvalho ATD, Cândido EB, Furtado RDS, Silva Filho ALD. Systemic Inflammatory Patterns in Ovarian Cancer Patients: Analysis of Cytokines, Chemokines, and Microparticles. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2023; 45:e780-e789. [PMID: 38141599 DOI: 10.1055/s-0043-1772590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2023] Open
Abstract
OBJECTIVE To compare the patterns of systemic inflammatory response in women with epithelial ovarian cancer (EOC) or no evidence of malignant disease, as well as to evaluate the profile of systemic inflammatory responses in type-1 and type-2 tumors. This is a non-invasive and indirect way to assess both tumor activity and the role of the inflammatory pattern during pro- and antitumor responses. MATERIALS AND METHODS We performed a prospective evaluation of 56 patients: 30 women without evidence of malignant disease and 26 women with EOC. The plasma quantification of cytokines, chemokines, and microparticles (MPs) was performed using flow cytometry. RESULTS Plasma levels of proinflammatory cytokines interleukin-12 (IL12), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) interleukin-1 beta (IL-1β), and interleukin-10 (IL-10), and C-X-C motif chemokine ligand 9 (CXCL-9) and C-X-C motif chemokine ligand 10 (CXCL-10) were significantly higher in patients with EOC than in those in the control group. Plasma levels of cytokine interleukin-17A (IL-17A) and MPs derived from endothelial cells were lower in patients with EOC than in the control group. The frequency of leukocytes and MPs derived from endothelial cells was higher in type-2 tumors than in those without malignancy. We observed an expressive number of inflammatory/regulatory cytokines and chemokines in the cases of EOC, as well as negative and positive correlations involving them, which leads to a higher complexity of these networks. CONCLUSION The present study showed that, through the development of networks consisting of cytokines, chemokines, and MPs, there is a greater systemic inflammatory response in patients with EOC and a more complex correlation of these biomarkers in type-2 tumors.
Collapse
Affiliation(s)
- Aline Evangelista Santiago
- Department of Gynecology and Obstetrics, Faculty of Medicine, Universidade Estadual Paulista "Júlio de Mesquita Filho", Botucatu, SP, Brasil
| | - Sálua Oliveira Calil de Paula
- Department of Gynecology and Obstetrics, Faculty of Medicine, Universidade Estadual Paulista "Júlio de Mesquita Filho", Botucatu, SP, Brasil
| | | | - Eduardo Batista Cândido
- Department of Gynecology and Obstetrics, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Rafaela de Souza Furtado
- Department of Gynecology and Obstetrics, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Agnaldo Lopes da Silva Filho
- Department of Gynecology and Obstetrics, Faculty of Medicine, Universidade Estadual Paulista "Júlio de Mesquita Filho", Botucatu, SP, Brasil
| |
Collapse
|
19
|
Phung MT, Lee AW, McLean K, Anton-Culver H, Bandera EV, Carney ME, Chang-Claude J, Cramer DW, Doherty JA, Fortner RT, Goodman MT, Harris HR, Jensen A, Modugno F, Moysich KB, Pharoah PDP, Qin B, Terry KL, Titus LJ, Webb PM, Wu AH, Zeinomar N, Ziogas A, Berchuck A, Cho KR, Hanley GE, Meza R, Mukherjee B, Pike MC, Pearce CL, Trabert B. A framework for assessing interactions for risk stratification models: the example of ovarian cancer. J Natl Cancer Inst 2023; 115:1420-1426. [PMID: 37436712 PMCID: PMC10637032 DOI: 10.1093/jnci/djad137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/08/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Generally, risk stratification models for cancer use effect estimates from risk/protective factor analyses that have not assessed potential interactions between these exposures. We have developed a 4-criterion framework for assessing interactions that includes statistical, qualitative, biological, and practical approaches. We present the application of this framework in an ovarian cancer setting because this is an important step in developing more accurate risk stratification models. Using data from 9 case-control studies in the Ovarian Cancer Association Consortium, we conducted a comprehensive analysis of interactions among 15 unequivocal risk and protective factors for ovarian cancer (including 14 non-genetic factors and a 36-variant polygenic score) with age and menopausal status. Pairwise interactions between the risk/protective factors were also assessed. We found that menopausal status modifies the association among endometriosis, first-degree family history of ovarian cancer, breastfeeding, and depot-medroxyprogesterone acetate use and disease risk, highlighting the importance of understanding multiplicative interactions when developing risk prediction models.
Collapse
Affiliation(s)
- Minh Tung Phung
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Alice W Lee
- Department of Public Health, California State University, Fullerton, Fullerton, CA, USA
| | - Karen McLean
- Department of Gynecologic Oncology and Department of Pharmacology & Therapeutics, Elm & Carlton Streets, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Hoda Anton-Culver
- Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Elisa V Bandera
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Michael E Carney
- Department of Obstetrics and Gynecology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel W Cramer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer Anne Doherty
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Renee T Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Allan Jensen
- Department of Lifestyle, Reproduction and Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Francesmary Modugno
- Women’s Cancer Research Center, Magee-Women’s Research Institute and Hillman Cancer Center, Pittsburgh, PA, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburg, PA, USA
| | - Kirsten B Moysich
- Division of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Paul D P Pharoah
- Department of Computational Biomedicine, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - Bo Qin
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kathryn L Terry
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Linda J Titus
- Public Health, Muskie School of Public Service, University of Southern Maine, Portland, ME, USA
| | - Penelope M Webb
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nur Zeinomar
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Argyrios Ziogas
- Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Andrew Berchuck
- Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Kathleen R Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gillian E Hanley
- Department of Obstetrics & Gynecology, University of British Columbia Faculty of Medicine, Vancouver, BC, Canada
| | - Rafael Meza
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Bhramar Mukherjee
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Malcolm C Pike
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Britton Trabert
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
- Cancer Control and Populations Sciences Program, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
20
|
Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2023; 49:2563-2574. [PMID: 37565583 DOI: 10.1111/jog.15761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
AIM This review aimed to describe the potential for therapeutic targeting of the JAK/STAT signaling pathway by repurposing the clinically-approved JAK inhibitor ruxolitinib in the patients with epithelial ovarian cancer (OC) setting. METHODS We reviewed publications that focus on the inhibition of the JAK/STAT pathway in hematological and solid malignancies including OC. RESULTS Preclinical studies showed that ruxolitinib effectively reduces OC cell viability and metastasis and enhances the anti-tumor activity of chemotherapy drugs. There are a number of recent clinical trials exploring the role of JAK/STAT inhibition in solid cancers including OC. Early results have not adequately supported efficacy in solid tumors. However, there are preclinical data and clinical studies supporting the use of ruxolitinib in combination with both chemotherapy and other targeted drugs in OC setting. CONCLUSION Inflammatory conditions and persistent activation of the JAK/STAT pathway are associated with tumourigenesis and chemoresistance, and therapeutic blockade of this pathway shows promising results. For women with OC, clinical investigation exploring the role of ruxolitinib in combination with chemotherapy agents or other targeted therapeutics is warranted.
Collapse
Affiliation(s)
- Irfan Yunianto
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
- Department of Biology Education, Universitas Ahmad Dahlan, Indonesia
| | - Margaret Currie
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
21
|
Jahani S, Zare N, Mirzaei Y, Arefnezhad R, Zarei H, Goleij P, Bagheri N. Mesenchymal stem cells and ovarian cancer: Is there promising news? J Cell Biochem 2023; 124:1437-1448. [PMID: 37682985 DOI: 10.1002/jcb.30471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023]
Abstract
Ovarian cancer (OC) is described as a heterogeneous complex condition with high mortality, weak prognosis, and late-stage presentation. OC has several subgroups based on different indices, like the origin and histopathology. The current treatments against OC include surgery followed by chemotherapy and radiotherapy; however, these methods have represented diverse side effects without enough effectiveness on OC. Recently, mesenchymal stem cell (MSC)-based therapy has acquired particular attention for treating diverse problems, such as cancer. These multipotent stem cells can be obtained from different sources, such as the umbilical cord, adipose tissues, bone marrow, and placenta, and their efficacy has been investigated against OC. Hence, in this narrative review, we aimed to review and discuss the present studies about the effects of various sources of MSCs on OC with a special focus on involved mechanisms.
Collapse
Affiliation(s)
| | - Nabi Zare
- Coenzyme R Research Institute, Tehran, Iran
| | - Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | | | - Hooman Zarei
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Sana Institute of Higher Education, Sari, Iran
- International Network of Stem Cell (INSC), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
22
|
Afshari H, Noori S, Zarghi A. Curcumin potentiates the anti-inflammatory effects of Tehranolide by modulating the STAT3/NF-κB signaling pathway in breast and ovarian cancer cell lines. Inflammopharmacology 2023; 31:2541-2555. [PMID: 37452228 DOI: 10.1007/s10787-023-01281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Studies have demonstrated that natural products, such as curcumin and artemisinin, possess anti-inflammatory effects, which can be beneficial for cancer treatment. Tehranolide, as a novel natural product, has a wide range of biological activities, including anti-cancer effects. However, many properties of Tehranolide, like its anti-inflammatory activity and its combination with curcumin, have not been investigated yet. This investigation examined the anti-inflammatory activity of Tehranolide, either alone or in combination with curcumin, via modulating the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and STAT3 (signal transducer and activator of transcription 3) signaling pathways in MDA-MB-231 and SKOV3, breast and ovarian cancer cell lines. METHODS ELISA-based methods were employed to measure the pro-inflammatory cytokine levels and the NF-κB activity in lipopolysaccharide (LPS)-induced cells. The real-time PCR experiment and Griess test were performed to evaluate inducible nitric oxide synthase (iNOS) gene expression and nitrite levels, respectively. The STAT3 and NF-κB signaling pathways were investigated by Western blotting analysis. Tehranolide's anti-cancer activity was also assessed in a mouse model of breast cancer using the TUNEL (terminal deoxynucleotidyl transferase nick-end labeling) assay. RESULTS Tehranolide diminished levels of pro-inflammatory cytokines in cancer cells. Additionally, it suppressed NF-κB DNA binding and STAT3 phosphorylation, reducing iNOS gene expression and nitrite production. Moreover, Western blotting showed that Tehranolide enhanced the inhibitory κB (IκBα) and Bcl-2 (B-cell lymphoma 2)-associated X (BAX) expression, and downregulated the expression of Bcl-2 proteins. Furthermore, the TUNEL assay demonstrated that Tehranolide induced apoptosis in a breast cancer mouse model. Curcumin potentiated all the anti-inflammatory effects of Tehranolide. CONCLUSION This investigation indicated for the first time that Tehranolide, either alone or in combination with curcumin, exerted its anti-inflammatory effects by suppressing NF-κB and STAT3 signaling pathways in SKOV3 and MDA-MB-231 cells.
Collapse
Affiliation(s)
- Havva Afshari
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Li M, Yan Y, Liu Y, Zhao J, Guo F, Chen J, Nie L, Zhang Y, Wang Y. Comprehensive analyses of fatty acid metabolism-related lncRNA for ovarian cancer patients. Sci Rep 2023; 13:14675. [PMID: 37673886 PMCID: PMC10482851 DOI: 10.1038/s41598-023-35218-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/15/2023] [Indexed: 09/08/2023] Open
Abstract
Ovarian cancer (OC) is a disease with difficult early diagnosis and treatment and poor prognosis. OC data profiles were downloaded from The Cancer Genome Atlas. Eight key fatty acid metabolism-related long non-coding RNAs (lncRNAs) were finally screened for building a risk scoring model by univariate/ multifactor and least absolute shrinkage and selection operator (LASSO) Cox regression. To make this risk scoring model more applicable to clinical work, we established a nomogram containing the clinical characteristics of OC patients after confirming that the model has good reliability and validity and the ability to distinguish patient prognosis. To further explore how these key lncRNAs are involved in OC progression, we explored their relationship with LUAD immune signatures and tumor drug resistance. The structure shows that the risk scoring model established based on these 8 fatty acid metabolism-related lncRNAs has good reliability and validity and can better predict the prognosis of patients with different risks of OC, and LINC00861in these key RNAs may be a hub gene that affects the progression of OC and closely related to the sensitivity of current OC chemotherapy drugs. In addition, combined with immune signature analysis, we found that patients in the high-risk group are in a state of immunosuppression, and Tfh cells may play an important role in it. We innovatively established a prognostic prediction model with excellent reliability and validity from the perspective of OC fatty acid metabolism reprogramming and lncRNA regulation and found new molecular/cellular targets for future OC treatment.
Collapse
Affiliation(s)
- Min Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Gynecology, Jincheng People's Hospital, Jincheng, 048026, China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yanyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jianzhen Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Fei Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jianqin Chen
- Department of Gynecology, Jincheng People's Hospital, Jincheng, 048026, China
| | - Lifang Nie
- Department of Gynecology, Jincheng People's Hospital, Jincheng, 048026, China
| | - Yong Zhang
- Department of Pathology, Jincheng People's Hospital, Jincheng, 048026, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping, Tianjin, 300052, China.
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
24
|
Afshari H, Noori S, Nourbakhsh M, Daraei A, Azami Movahed M, Zarghi A. A novel imidazo[1,2-a]pyridine derivative and its co-administration with curcumin exert anti-inflammatory effects by modulating the STAT3/NF-κB/iNOS/COX-2 signaling pathway in breast and ovarian cancer cell lines. BIOIMPACTS : BI 2023; 14:27618. [PMID: 38505673 PMCID: PMC10945297 DOI: 10.34172/bi.2023.27618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/15/2023] [Accepted: 04/26/2023] [Indexed: 03/21/2024]
Abstract
Introduction Imidazo[1,2-a]pyridine derivatives with diverse pharmacological properties and curcumin, as a potential natural anti-inflammatory compound, are promising compounds for cancer treatment. This study aimed to synthesize a novel imidazo[1,2-a]pyridine derivative, (MIA), and evaluate its anti-inflammatory activity and effects on nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways, and their target genes, alone and in combination with curcumin, in MDA-MB-231 and SKOV3 cell lines. Methods We evaluated the interaction between imidazo[1,2-a]pyridine ligand, curcumin, and NF-κB p50 protein, using molecular docking studies. MTT assay was used to investigate the impacts of compounds on cell viability. To evaluate the NF-κB DNA binding activity and the level of inflammatory cytokines in response to the compounds, ELISA-based methods were performed. In addition, quantitative polymerase chain reaction (qPCR) and western blotting were carried out to analyze the expression of genes and investigate NF-κB and STAT3 signaling pathways. Results Molecular docking studies showed that MIA docked into the NF-κB p50 subunit, and curcumin augmented its binding. The MTT assay results indicated that MIA and its combination with curcumin reduced cell viability. According to the results of the ELISA-based methods, MIA lowered the levels of inflammatory cytokines and suppressed NF-κB activity. In addition, real-time PCR and Griess test results showed that the expression of cyclooxygenase-2 (COX-2) and inducible NO synthase (iNOS) genes, and nitrite production were reduced by MIA. Furthermore, the western blotting analysis demonstrated that MIA increased the expression of inhibitory κB (IκBα) and B-cell lymphoma 2 (Bcl-2)-associated X proteins (BAX), and suppressed the STAT3 phosphorylation, and Bcl-2 expression. Our findings revealed that curcumin had a potentiating role and enhanced all the anti-inflammatory effects of MIA. Conclusion This study indicated that the anti-inflammatory activity of MIA is exerted by suppressing the NF-κB and STAT3 signaling pathways in MDA-MB-231 and SKOV3 cancer cell lines.
Collapse
Affiliation(s)
- Havva Afshari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azam Daraei
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Azami Movahed
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Huang M, Wu Z, Jia L, Wang Y, Gao S, Liu Y, Zhang Y, Li J. Bioinformatics and network pharmacology identify promotional effects and potential mechanisms of ethanol on esophageal squamous cell carcinoma and experimental validation. Toxicol Appl Pharmacol 2023; 474:116615. [PMID: 37406968 DOI: 10.1016/j.taap.2023.116615] [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: 04/06/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Ethanol is an important risk factor for esophageal squamous cell carcinoma (ESCC); however, the molecular mechanisms behind how ethanol promotes ESCC development remain poorly understood. In this study, ethanol-ESCC-associated target genes were constructed and screened using network pharmacology and subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) and bioinformatics analysis. A mouse ethanol-exposed esophageal cancer model was constructed with 4-nitroquinoline-1-oxide (4-NQO) to assess its survival and tumor lesion status, and the mechanism of ethanol-promoted ESCC lesions was verified by qRT-PCR and Western blotting. The results showed that 126 ethanol-ESCC crossover genes were obtained, which were significantly enriched in the PI3K/AKT signaling pathway. Bioinformatics results showed that the target genes TNF, IL6, IL1β and JUN were highly expressed in esophageal tumor samples and positively correlated with tumor proliferation and apoptosis genes, and the genetic information of these genes was mutated to different degrees. Animal model experiments showed that ethanol decreased the survival rate and aggravated the occurrence of esophageal cancer in mice. qRT-PCR showed that ethanol promoted the expression of TNF, IL6, IL1β and JUN mRNA in mouse esophageal tumor tissues, and Western blotting showed that ethanol promoted p-PI3K and p-AKT protein expression in mouse esophageal tumor tissues. In conclusion, ethanol promotes esophageal carcinogenesis by increasing the expression of TNF, IL6, IL1β and JUN and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhongbing Wu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Jia
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuang Gao
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Liu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yushuang Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China; The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| |
Collapse
|
26
|
Guo JY, Wang YJ, Li SQ, Wu YP. Molecular targets of metformin against ovarian cancer based on network pharmacology. Chem Biol Drug Des 2023; 102:88-100. [PMID: 36977503 DOI: 10.1111/cbdd.14234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/23/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
The objective of this study was to analyze potential targets of metformin against ovarian cancer (OC) through network pharmacology. Pharmacodynamic targets of metformin were predicted using the Bioinformatics Analysis Tool for the molecular mechanism of traditional Chinese medicine (BATMAN), Drugbank, PharmMapper, SwissTargetPrediction, and TargetNet databases. R was utilized to analyze the gene expression of OC tissues, normal/adjacent noncancerous tissues, and screen differentially expressed genes (DEGs) in the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) + Genotype-Tissue Expression (GTEx) datasets. STRING 11.0 was utilized to explore the protein-protein interaction (PPI) of metformin target genes differentially expressed in OC. Cytoscape 3.8.0 was used to construct the network and screen the core targets. Additionally, gene ontology (GO) annotation and enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed for the common targets of metformin and OC through the DAVID 6.8 database. A total of 95 potential common targets of metformin and OC were identified from the intersection of 255 potential pharmacodynamic targets of metformin and 10,463 genes associated with OC. Furthermore, 10 core targets were screened from the PPI network [e.g., interleukin (IL) 1B, KCNC1, ESR1, HTR2C, MAOB, GRIN2A, F2, GRIA2, APOE, PTPRC]. In addition, it was shown in GO enrichment analysis that the common targets were mainly associated with biological processes (i.e., response to stimuli or chemical, cellular processes, and transmembrane transport), cellular components (i.e., plasma membrane, cell junction, and cell projection), and molecular functions (i.e., binding, channel activities, transmembrane transporter activity, and signaling receptor activities). Furthermore, it was indicated by KEGG pathway analysis that the common targets were enriched in metabolic pathways. The critical molecular targets and molecular pathways of metformin against OC were preliminarily determined by bioinformatics-based network pharmacology analysis, providing a basis, and reference for further experimental studies.
Collapse
Affiliation(s)
- Jia-Yi Guo
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Beijing, China
| | - Yong-Jun Wang
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Beijing, China
| | - Si-Qi Li
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Beijing, China
| | - Yu-Ping Wu
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
27
|
Garg P, Awasthi S, Horne D, Salgia R, Singhal SS. The innate effects of plant secondary metabolites in preclusion of gynecologic cancers: Inflammatory response and therapeutic action. Biochim Biophys Acta Rev Cancer 2023; 1878:188929. [PMID: 37286146 DOI: 10.1016/j.bbcan.2023.188929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Gynecologic cancers can make up the bulk of cancers in both humans and animals. The stage of diagnosis and the type of tumor, its origin, and its spread are a few of the factors that influence how effectively a treatment modality works. Currently, radiotherapy, chemotherapy, and surgery are the major treatment options recommended for the eradication of malignancies. The use of several anti-carcinogenic drugs increases the chance of harmful side effects, and patients might not react to the treatments as expected. The significance of the relationship between inflammation and cancer has been underscored by recent research. As a result, it has been shown that a variety of phytochemicals with beneficial bioactive effects on inflammatory pathways have the potential to act as anti-carcinogenic medications for the treatment of gynecologic cancer. The current paper reviews the significance of inflammatory pathways in gynecologic malignancies and discusses the role of plants-derived secondary metabolites that are useful in the treatment of cancer.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sanjay Awasthi
- Cayman Health, CTMH Doctors Hospital in Cayman Islands, George Town, Grand Cayman, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
28
|
Liu W, Wang L, Zhang J, Cheng K, Zheng W, Ma Z. CC Chemokine 2 Promotes Ovarian Cancer Progression through the MEK/ERK/MAP3K19 Signaling Pathway. Int J Mol Sci 2023; 24:10652. [PMID: 37445830 DOI: 10.3390/ijms241310652] [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: 05/23/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer is a gynecological tumor with an incidence rate lower than those of other gynecological tumor types and the second-highest death rate. CC chemokine 2 (CCL2) is a multifunctional factor associated with the progression of numerous cancers. However, the effect of CCL2 on ovarian cancer progression is unclear. Here, we found that exogenous CCL2 and the overexpression of CCL2 promoted the proliferation and metastasis of ovarian cancer cells. On the other hand, CCL2 knockdown via CRISPR/Cas9 inhibited ovarian cancer cell proliferation, migration, and invasion. The present study demonstrated that mitogen-activated protein three kinase 19 (MAP3K19) was the key CCL2 target for regulating ovarian cancer progression through transcriptome sequencing. Additionally, MAP3K19 knockout inhibited ovarian cancer cell proliferation, migration, and invasion. Furthermore, CCL2 increased MAP3K19 expression by activating the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. The present study showed the correlation between CCL2 and ovarian cancer, suggesting that CCL2 may be a novel target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Lei Wang
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Jiajia Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Kun Cheng
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenming Zheng
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Zhenling Ma
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
29
|
Dong L, Qian YP, Li SX, Pan H. Development of a machine learning-based signature utilizing inflammatory response genes for predicting prognosis and immune microenvironment in ovarian cancer. Open Med (Wars) 2023; 18:20230734. [PMID: 37273921 PMCID: PMC10238811 DOI: 10.1515/med-2023-0734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023] Open
Abstract
Ovarian cancer (OC) represents a significant health challenge, characterized by a particularly unfavorable prognosis for affected women. Accumulating evidence supports the notion that inflammation-related factors impacting the normal ovarian epithelium may contribute to the development of OC. However, the precise role of inflammatory response-related genes (IRRGs) in OC remains largely unknown. To address this gap, we performed an integration of mRNA expression profiles from 7 cohorts and conducted univariate Cox regression analysis to screen 26 IRRGs. By utilizing these IRRGs, we categorized patients into subtypes exhibiting diverse inflammatory responses, with subtype B displaying the most prominent immune infiltration. Notably, the elevated abundance of Treg cells within subtype B contributed to immune suppression, resulting in an unfavorable prognosis for these patients. Furthermore, we validated the distribution ratios of stromal cells, inflammatory cells, and tumor cells using whole-slide digitized histological slides. We also elucidated differences in the activation of biological pathways among subtypes. In addition, machine learning algorithms were employed to predict the likelihood of survival in OC patients based on the expression of prognostic IRRGs. Through rigorous testing of over 100 combinations, we identified CXCL10 as a crucial IRRG. Single-cell analysis and vitro experiments further confirmed the potential secretion of CXCL10 by macrophages and its involvement in lymphangiogenesis within the tumor microenvironment. Overall, the study provides new insights into the role of IRRGs in OC and may have important implications for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Li Dong
- Department of Obstetrics and Gynaecology, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, No. 7 People’s Hospital, Changzhou, China
| | - Ya-ping Qian
- Department of Obstetrics and Gynaecology, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, No. 7 People’s Hospital, Changzhou, China
| | - Shu-xiu Li
- Department of Obstetrics and Gynaecology, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, No. 7 People’s Hospital, Changzhou, China
| | - Hao Pan
- Department of Cardiology, The Affiliated Changzhou, No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
30
|
Choi WK, Hussein UK, Ahmed AG, Zhang J, Kim KM, Ahn AR, Park HS, Park SH, Cho DH, Jang KY. Co-operative roles of IL-4Rα and IL-13Rα1 in the progression of ovarian carcinomas and the survival of ovarian carcinoma patients. Cancer Commun (Lond) 2023. [PMID: 37243576 PMCID: PMC10354408 DOI: 10.1002/cac2.12445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/16/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023] Open
Affiliation(s)
- Won Ku Choi
- Department of Obstetrics and Gynecology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Usama Khamis Hussein
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Asmaa Gamal Ahmed
- Faculty of Postgraduate Studies and Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Junyue Zhang
- Department of Orthopedic Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Ae Ri Ahn
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Ho Sung Park
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Dong Hyu Cho
- Department of Obstetrics and Gynecology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
31
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
32
|
Asp M, Mockute I, Måsbäck A, Liuba K, Kannisto P, Malander S. Tru-Cut Biopsy in Gynecological Cancer: Adequacy, Accuracy, Safety and Clinical Applicability. J Multidiscip Healthc 2023; 16:1367-1377. [PMID: 37215751 PMCID: PMC10198176 DOI: 10.2147/jmdh.s396788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Purpose Tru-cut biopsy is a minimally invasive technique used to obtain tissue samples for the diagnosis of tumors, especially in patients where primary surgery is not indicated. The aim of this study was to assess the adequacy, accuracy and safety of the tru-cut biopsy for diagnosis in gynecological cancer. Methods A retrospective population-based review of 328 biopsies was conducted. The indications for tru-cut biopsies were diagnosis of primary tumors, metastases of gynecological and non-gynecological tumors, and suspected recurrences. A tissue sample was considered adequate when the quality/quality was sufficient to identify the subtype/origin of the tumor. Potential factors affecting adequacy were analyzed using logistic regressions analyses. Accuracy was defined as agreement between the diagnosis of the tru-cut biopsy and the postoperative histology. The therapy plan was registered, and the clinical applicability of the tru-cut biopsy was investigated. Complications within 30 days after the biopsy procedure were registered. Results In total, 300 biopsies were identified as tru-cut biopsies. The overall adequacy was 86.3%, varying between 80.8% and 93.5%, respectively, when performed by a gynecological oncologist or a gynecologist with a subspecialty in ultrasound diagnosis. Sampling of a pelvic mass had a lower adequacy (81.6%) compared with sampling of the omentum (93.9%) or carcinomatosis (91.5%). The accuracy was 97.5%, and the complication rate was 1.3%. Conclusion The tru-cut biopsy is a safe and reliable diagnostic method with a high accuracy and a good adequacy, depending on the site of the tissue sample, indications for the biopsy and the experience of the operator.
Collapse
Affiliation(s)
- Mihaela Asp
- Department of Obstetrics and Gynecology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Ingrida Mockute
- Department of Obstetrics and Gynecology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Anna Måsbäck
- Department of Clinical Genetics and Pathology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Karina Liuba
- Department of Obstetrics and Gynecology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Päivi Kannisto
- Department of Obstetrics and Gynecology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Susanne Malander
- Department of Oncology and Pathology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Kwolek DG, Gerstberger S, Tait S, Qiu JM. Ovarian, Uterine, and Vulvovaginal Cancers: Screening, Treatment Overview, and Prognosis. Med Clin North Am 2023; 107:329-355. [PMID: 36759101 DOI: 10.1016/j.mcna.2022.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ovarian, uterine, and vulvovaginal cancers affect approximately 96,000 women per year in the United States, resulting in approximately 29,000 deaths annually. Routine screening protocols do not detect these malignancies; thus, the recognition of risk factors and evaluation of worrisome symptoms are essential for early detection and improved prognoses. Treatment is managed by gynecologic oncologists, and often involves a combination of surgery, chemotherapy, and possible radiation treatments. Survivor care is managed by the primary-care clinician: expert attention to the mental, physical, and sexual health of each patient will ensure the best outcomes and quality of life.
Collapse
Affiliation(s)
- Deborah Gomez Kwolek
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Stefanie Gerstberger
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sarah Tait
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Jeanna M Qiu
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
34
|
Kłysiak M, Wieder-Huszla S, Branecka-Woźniak D, Karakiewicz-Krawczyk K, Napieracz-Trzosek I, Owsianowska J, Jurczak A, Cymbaluk-Płoska A. Analysis of the Occurrence of Predicative Factors of Chronic Fatigue in Female Patients with Cancer of the Reproductive Organs with Respect to Stage of Treatment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3732. [PMID: 36834426 PMCID: PMC9967751 DOI: 10.3390/ijerph20043732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 06/18/2023]
Abstract
The aim of this study was to search for mechanisms contributing to cancer-related fatigue in patients with gynecologic cancer. The study involved 51 women with advanced endometrial cancer and ovarian cancer undergoing chemotherapy. Data were gathered at four points in time. After giving consent, each of the women had their blood drawn several times (before surgery and the first, third, and sixth cycle of chemotherapy) to determine serum levels of pro- and anti-inflammatory cytokines. Empirical data were collected using the MFSI-SF and an original questionnaire. Cancer-related fatigue (CRF) was present at every stage of treatment, but the highest mean scores were noted before cytoreductive surgery (8.745 ± 4.599), and before the sixth cycle of chemotherapy (9.667 ± 4.493). Statistically significant relationships were found between IL-1α, IL-1β, IL-2, Il-6, and IL-10 and fatigue at different stages of treatment. Older age and an above-normal BMI were the major prerequisite factors for the occurrence of fatigue in female oncological patients. The analysis of changes in cytokine levels and the severity of fatigue may be used to improve our understanding of cancer-related fatigue, and to take action to alleviate the obtrusive symptoms experienced by female patients with cancer of the reproductive organs.
Collapse
Affiliation(s)
- Magdalena Kłysiak
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Sylwia Wieder-Huszla
- Department of Clinical Nursing, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Dorota Branecka-Woźniak
- Department of Gynecology and Reproductive Health Pomeranian Medical University of Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | | | - Izabela Napieracz-Trzosek
- Department of Clinical Nursing, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Joanna Owsianowska
- Department of Clinical Nursing, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Anna Jurczak
- Department of Clinical Nursing, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
35
|
Xu S, Gao X, Qiu J, Hong F, Gao F, Wang X, Zhang S. TIPE2 acts as a tumor suppressor and correlates with tumor microenvironment immunity in epithelial ovarian cancer. Aging (Albany NY) 2023; 15:1052-1073. [PMID: 36801818 PMCID: PMC10008487 DOI: 10.18632/aging.204529] [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: 11/09/2022] [Accepted: 02/11/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is one of the deadliest gynecologic cancers. The etiology of EOC has still not been elucidated thoroughly. Tumor necrosis factor-α-induced protein 8-like2 (TNFAIP8L2, TIPE2), an important regulator of inflammation and immune homeostasis, plays a critical role in the progression of various cancers. This study aims to investigate the role of TIPE2 in EOC. METHODS Expression of TIPE2 protein and mRNA in EOC tissues and cell lines was examined using Western blot and quantitative real-time PCR (qRT-PCR). The functions of TIPE2 in EOC were investigated by cell proliferation assay, colony assay, transwell assay, and apoptosis analysis in vitro. To further investigate the regulatory mechanisms of TIPE2 in EOC, RNA-seq and western blot were performed. Finally, the CIBERSORT algorithm and databases including Tumor Immune Single-cell Hub (TISCH), Tumor Immune Estimation Resource (TIMER), Tumor-Immune System Interaction (TISIDB), and The Gene Expression Profiling Interactive Analysis (GEPIA) were used to elucidate its potential role in regulating tumor immune infiltration in the tumor microenvironment (TME). RESULTS TIPE2 expression was shown to be considerably lower in both EOC samples and cell lines. Overexpression of TIPE2 suppressed EOC cell proliferation, colony formation, and motility in vitro. Mechanistically, TIPE2 suppressed EOC by blocking the PI3K/Akt signaling pathway, according to bioinformatics analysis and western blot in TIPE2 overexpression EOC cell lines, and the anti-oncogenic potentials of TIPE2 in EOC cells could be partially abrogated by the PI3K agonist, 740Y-P. Finally, TIPE2 expression was positively associated with various immune cells and possibly involved in the regulation of macrophage polarization in ovarian cancer. CONCLUSIONS We detail the regulatory mechanism of TIPE2 in EOC carcinogenesis, as well as how it correlates with immune infiltration, emphasizing its potential as a therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Shuai Xu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China.,Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Xiaolin Gao
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Jianqing Qiu
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Fanzhen Hong
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Fufeng Gao
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xia Wang
- Laboratory of Translational Gastroenterology, Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| |
Collapse
|
36
|
Madeddu C, Busquets S, Donisi C, Lai E, Pretta A, López-Soriano FJ, Argilés JM, Scartozzi M, Macciò A. Effect of Cancer-Related Cachexia and Associated Changes in Nutritional Status, Inflammatory Status, and Muscle Mass on Immunotherapy Efficacy and Survival in Patients with Advanced Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:1076. [PMID: 36831431 PMCID: PMC9953791 DOI: 10.3390/cancers15041076] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Immune checkpoint inhibitor (ICI)-based immunotherapy has significantly improved the survival of patients with advanced non-small cell lung cancer (NSCLC); however, a significant percentage of patients do not benefit from this approach, and predictive biomarkers are needed. Increasing evidence demonstrates that cachexia, a complex syndrome driven by cancer-related chronic inflammation often encountered in patients with NSCLC, may impair the immune response and ICI efficacy. Herein, we carried out a prospective study aimed at evaluating the prognostic and predictive role of cachexia with the related changes in nutritional, metabolic, and inflammatory parameters (assessed by the multidimensional miniCASCO tool) on the survival and clinical response (i.e., disease control rate) to ICI-based immunotherapy in patients with advanced NSCLC. We included 74 consecutive patients. Upon multivariate regression analysis, we found a negative association between IL-6 levels (odds ratio (OR) = 0.9036; 95%CI = 0.8408-0.9711; p = 0.0025) and the miniCASCO score (OR = 0.9768; 95%CI = 0.9102-0.9999; p = 0.0310) with the clinical response. As for survival outcomes, multivariate COX regression analysis found that IL-6 levels and miniCASCO-based cachexia severity significantly affected PFS (hazard ratio (HR) = 1.0388; 95%CI = 1.0230-1.0548; p < 0.001 and HR = 1.2587; 95%CI = 1.0850-1.4602; p = 0.0024, respectively) and OS (HR = 1.0404; 95%CI = 1.0221-1.0589; p < 0.0001 and HR = 2.3834; 95%CI = 1.1504-4.9378; p = 0.0194, respectively). A comparison of the survival curves by Kaplan-Meier analysis showed a significantly lower OS in patients with cachexia versus those without cachexia (p = 0.0323), as well as higher miniCASCO-based cachexia severity (p = 0.0428), an mGPS of 2 versus those with a lower mGPS (p = 0.0074), and higher IL-6 levels (>6 ng/mL) versus those with lower IL-6 levels (≤6 ng/mL) (p = 0.0120). In conclusion, our study supports the evidence that cachexia, with its related changes in inflammatory, body composition, and nutritional parameters, is a key prognostic and predictive factor for ICIs. Further larger studies are needed to confirm these findings and to explore the potential benefit of counteracting cachexia to improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Clelia Madeddu
- Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| | - Silvia Busquets
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Clelia Donisi
- Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| | - Eleonora Lai
- Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| | - Andrea Pretta
- Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| | - Francisco Javier López-Soriano
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Josep Maria Argilés
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Mario Scartozzi
- Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| | - Antonio Macciò
- Gynecologic Oncology Unit, ARNAS G. Brotzu, Department of Surgical Sciences, University of Cagliari, 09100 Cagliari, Italy
| |
Collapse
|
37
|
Kampan NC, Kartikasari AER, Deceneux C, Madondo MT, McNally OM, Flanagan KL, Aziz NA, Stephens AN, Reynolds J, Quinn MA, Plebanski M. Combining TNFR2-Expressing Tregs and IL-6 as Superior Diagnostic Biomarkers for High-Grade Serous Ovarian Cancer Masses. Cancers (Basel) 2023; 15:667. [PMID: 36765633 PMCID: PMC9913655 DOI: 10.3390/cancers15030667] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
We hypothesised that the inclusion of immunosuppressive and inflammatory biomarkers in HGSOC patients would improve the sensitivity and specificity of the preoperative marker prediction of malignancy in patients with ovarian masses. We tested a panel of 29 soluble immune factors by multiplex bead immunoassay and 16 phenotypic T cell markers by flow cytometry in pre-treatment blood samples from 66 patients undergoing surgery for suspected ovarian cancer or ovarian cancer risk reduction. The potential diagnostic utility of all parameters was explored using Volcano plots, principal component analysis (PCA) and receiver operator characteristic (ROC) analysis. We also assessed the effect of culturing PBMCs from 20 healthy donors in the presence of malignant ascites fluid. The combination of TNFR2+ Tregs and IL-6 in the pre-treatment blood of patients with advanced HGSOC effectively discriminated patients with benign or malignant ovarian masses. In vitro culturing of the PBMCs of healthy donors in malignant ascites promoted an increase in TNFR2-expressing Tregs, which were decreased following blockade with IL-6 or STAT3 activity. Pre-treatment serum IL-6 and peripheral blood TNFR2+ Tregs may be potential clinical biomarkers that can discriminate patients with malignant compared to benign ovarian cancer masses, and the relationship between IL-6 and TNFR2+ Treg is likely to be mediated via the STAT3 signalling pathway.
Collapse
Affiliation(s)
- Nirmala Chandralega Kampan
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | | | - Cyril Deceneux
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Mutsa Tatenda Madondo
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Orla M. McNally
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Melbourne University, Parkville, VIC 3052, Australia
| | - Katie Louise Flanagan
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Hobart, TAS 7005, Australia
| | - Norhaslinda A. Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Andrew N. Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
- Epworth Research Institute, Epworth Healthcare, Richmond, VIC 3121, Australia
| | - John Reynolds
- Biostatistics Consulting Platform, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Michael A. Quinn
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
| | - Magdalena Plebanski
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| |
Collapse
|
38
|
Mo Y, Leung LL, Mak CSL, Wang X, Chan WS, Hui LMN, Tang HWM, Siu MKY, Sharma R, Xu D, Tsui SKW, Ngan HYS, Yung MMH, Chan KKL, Chan DW. Tumor-secreted exosomal miR-141 activates tumor-stroma interactions and controls premetastatic niche formation in ovarian cancer metastasis. Mol Cancer 2023; 22:4. [PMID: 36624516 PMCID: PMC9827705 DOI: 10.1186/s12943-022-01703-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Metastatic colonization is one of the critical steps in tumor metastasis. A pre-metastatic niche is required for metastatic colonization and is determined by tumor-stroma interactions, yet the mechanistic underpinnings remain incompletely understood. METHODS PCR-based miRNome profiling, qPCR, immunofluorescent analyses evaluated the expression of exosomal miR-141 and cell-to-cell communication. LC-MS/MS proteomic profiling and Dual-Luciferase analyses identified YAP1 as the direct target of miR-141. Human cytokine profiling, ChIP, luciferase reporter assays, and subcellular fractionation analyses confirmed YAP1 in modulating GROα production. A series of in vitro tumorigenic assays, an ex vivo model and Yap1 stromal conditional knockout (cKO) mouse model demonstrated the roles of miR-141/YAP1/GROα/CXCR1/2 signaling cascade. RNAi, CRISPR/Cas9 and CRISPRi systems were used for gene silencing. Blood sera, OvCa tumor tissue samples, and tissue array were included for clinical correlations. RESULTS Hsa-miR-141-3p (miR-141), an exosomal miRNA, is highly secreted by ovarian cancer cells and reprograms stromal fibroblasts into proinflammatory cancer-associated fibroblasts (CAFs), facilitating metastatic colonization. A mechanistic study showed that miR-141 targeted YAP1, a critical effector of the Hippo pathway, reducing the nuclear YAP1/TAZ ratio and enhancing GROα production from stromal fibroblasts. Stromal-specific knockout (cKO) of Yap1 in murine models shaped the GROα-enriched microenvironment, facilitating in vivo tumor colonization, but this effect was reversed after Cxcr1/2 depletion in OvCa cells. The YAP1/GROα correlation was demonstrated in clinical samples, highlighting the clinical relevance of this research and providing a potential therapeutic intervention for impeding premetastatic niche formation and metastatic progression of ovarian cancers. CONCLUSIONS This study uncovers miR-141 as an OvCa-derived exosomal microRNA mediating the tumor-stroma interactions and the formation of tumor-promoting stromal niche through activating YAP1/GROα/CXCRs signaling cascade, providing new insight into therapy for OvCa patients with peritoneal metastases.
Collapse
Affiliation(s)
- Yulan Mo
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Leanne L. Leung
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Celia S. L. Mak
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Xueyu Wang
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Wai-Sun Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Lynn M. N. Hui
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Hermit W. M. Tang
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Michelle K. Y. Siu
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Rakesh Sharma
- grid.194645.b0000000121742757Centre for PanorOmic Sciences Proteomics and Metabolomics Core, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Dakang Xu
- grid.16821.3c0000 0004 0368 8293Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Stephen K. W. Tsui
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, SAR Hong Kong, China
| | - Hextan Y. S. Ngan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Mingo M. H. Yung
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - Karen K. L. Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China
| | - David W. Chan
- grid.194645.b0000000121742757Department of Obstetrics & Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR China ,grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, SAR Hong Kong, China ,grid.511521.3School of Medicine, The Chinese University of Hong Kong-Shenzhen, Shenzhen, 518172 China
| |
Collapse
|
39
|
Hathaway CA, Wang T, Townsend MK, Vinci C, Jake-Schoffman DE, Saeed-Vafa D, Segura CM, Nguyen JV, Conejo-Garcia JR, Fridley BL, Tworoger SS. Lifetime Exposure to Cigarette Smoke and Risk of Ovarian Cancer by T-cell Tumor Immune Infiltration. Cancer Epidemiol Biomarkers Prev 2023; 32:66-73. [PMID: 36318652 PMCID: PMC9839509 DOI: 10.1158/1055-9965.epi-22-0877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/21/2022] [Accepted: 10/27/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Exposure to cigarette smoke, particularly in early life, is modestly associated with ovarian cancer risk and may impact systemic immunity and the tumor immune response. However, no studies have evaluated whether cigarette smoke exposure impacts the ovarian tumor immune microenvironment. METHODS Participants in the Nurses' Health Study (NHS) and NHSII reported on early life exposure to cigarette smoke and personal smoking history on questionnaires (n = 165,760). Multiplex immunofluorescence assays were used to measure markers of T cells and immune checkpoints in tumor tissue from 385 incident ovarian cancer cases. We used Cox proportional hazards models to evaluate HRs and 95% confidence intervals (CI) for developing ovarian tumors with a low (<median) or high (≥median) immune cell percentage by cigarette exposure categories. RESULTS Women exposed versus not to cigarette smoke early in life had a higher risk of developing ovarian cancer with low levels of T cells overall (CD3+: HR: 1.54, 95% CI: 1.08-2.20) and recently activated cytotoxic T cells (CD3+CD8+CD69+: HR: 1.45, 95% CI: 1.05-2.00). These findings were not statistically significant at the Bonferroni-corrected P value of 0.0083. Adult smoking was not significantly associated with tumor immune markers after Bonferroni correction. CONCLUSIONS These results suggest early life cigarette smoke exposure may modestly increase risk of developing ovarian tumors with low abundance of total T cells and recently activated cytotoxic T cells. IMPACT Future research should focus on understanding the impact of exposures throughout the life course on the ovarian tumor immune microenvironment.
Collapse
Affiliation(s)
| | - Tianyi Wang
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Mary K. Townsend
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Christine Vinci
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Daryoush Saeed-Vafa
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, Florida, USA.,Advanced Analytical and Digital Laboratory, Moffitt Cancer Center, Tampa, Florida, USA
| | - Carlos Moran Segura
- Advanced Analytical and Digital Laboratory, Moffitt Cancer Center, Tampa, Florida, USA
| | - Jonathan V. Nguyen
- Advanced Analytical and Digital Laboratory, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Brooke L. Fridley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, USA
| | - Shelley S. Tworoger
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
40
|
Singh RD, Dholariya S, Shekher A, Avadhesh, Parchwani D, Gupta SC. Role of IL-1 gene polymorphisms in common solid cancers. MULTIFACETED ROLE OF IL-1 IN CANCER AND INFLAMMATION 2023:1-69. [DOI: 10.1016/b978-0-12-824273-5.00002-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
41
|
Pan X, Shen Q, Zhang C, Zhang X, Li Y, Chang Z, Pang B. Coicis Semen for the treatment of malignant tumors of the female reproductive system: A review of traditional Chinese medicinal uses, phytochemistry, pharmacokinetics, and pharmacodynamics. Front Pharmacol 2023; 14:1129874. [PMID: 36909176 PMCID: PMC9995914 DOI: 10.3389/fphar.2023.1129874] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Coicis Semen is an important food product and traditional Chinese medicine (TCM) derived from the dried and mature seeds of Coix lacryma-jobi L.var.ma-yuen (Roman.) Stapf. An increasing number of studies have investigated its use, either alone or in combination with other botanical drugs, to treat female reproductive system malignancies, and its pharmacological effects have been confirmed clinically. This review aims to provide an overview of Coicis Semen's historical role in treating female reproductive system malignancies based on TCM theory, to summarize clinical trials results, and to analyze information pertaining to the main phytochemical components, pharmacokinetics, related anti-cancer pharmacological effects, and toxicology of Coicis Semen. Information on Coicis Semen was collected from internationally accepted scientific databases. Seventy-four clinical trials were identified that used Coicis Semen in combination with other Chinese medicine to treat female reproductive system malignancies, most of which demonstrated good anti-tumor efficacy and few adverse reactions. To date, more than 80 individual compounds have been isolated from this botanical drug. In terms of anti-tumor effects, Coix seed oil has been studied the most. Pharmacokinetic data suggest that the active ingredients in Coicis Semen are widely distributed after administration, and Coicis Semen and its active compounds play a beneficial role in treating female reproductive system malignancies. Mechanistically, the anti-cancer effects may be related to inhibition of tumor cell proliferation and promotion of apoptosis, inhibition of tumor angiogenesis, suppression of the chronic inflammatory microenvironment of tumors, modulation of immune function, and regulation of the female reproductive system. Most acute toxicity and genotoxicity studies have shown that Coicis Semen is non-toxic. However, the existing studies have many limitations, and the future research direction should emphasize 1) the relationship between drug concentration and pharmacological action as well as toxicity; 2) the structural modification or the synthesis of analogues led by the active ingredients of Coicis Semen to enhance pharmacological activities and bioavailability; 3) accurately revealing the anti-cancer pharmacological effects of Coicis Semen and its compounds through multi-omics technology. We hope that this review can determine future directions and inform novel drug development for treating female reproductive malignancies.
Collapse
Affiliation(s)
- Xue Pan
- Post-doctoral Mobile Station, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Shen
- International Medical Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanlong Zhang
- International Medical Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Yi Li
- International Medical Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuo Chang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Bo Pang
- International Medical Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Escalona RM, Chu S, Kadife E, Kelly JK, Kannourakis G, Findlay JK, Ahmed N. Knock down of TIMP-2 by siRNA and CRISPR/Cas9 mediates diverse cellular reprogramming of metastasis and chemosensitivity in ovarian cancer. Cancer Cell Int 2022; 22:422. [PMID: 36585738 PMCID: PMC9805260 DOI: 10.1186/s12935-022-02838-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The endogenous tissue inhibitor of metalloproteinase-2 (TIMP-2), through its homeostatic action on certain metalloproteinases, plays a vital role in remodelling extracellular matrix (ECM) to facilitate cancer progression. This study investigated the role of TIMP-2 in an ovarian cancer cell line in which the expression of TIMP-2 was reduced by either siRNA or CRISPR/Cas9. METHODS OVCAR5 cells were transiently and stably transfected with either single or pooled TIMP-2 siRNAs (T2-KD cells) or by CRISPR/Cas9 under the influence of two distinct guide RNAs (gRNA1 and gRNA2 cell lines). The expression of different genes was analysed at the mRNA level by quantitative real time PCR (qRT-PCR) and at the protein level by immunofluorescence (IF) and western blot. Proliferation of cells was investigated by 5-Ethynyl-2'-deoxyuridine (EdU) assay or staining with Ki67. Cell migration/invasion was determined by xCELLigence. Cell growth in vitro was determined by 3D spheroid cultures and in vivo by a mouse xenograft model. RESULTS Approximately 70-90% knock down of TIMP-2 expression were confirmed in T2-KD, gRNA1 and gRNA2 OVCAR5 ovarian cancer cells at the protein level. T2-KD, gRNA1 and gRNA2 cells exhibited a significant downregulation of MMP-2 expression, but concurrently a significant upregulation in the expression of membrane bound MMP-14 compared to control and parental cells. Enhanced proliferation and invasion were exhibited in all TIMP-2 knocked down cells but differences in sensitivity to paclitaxel (PTX) treatment were observed, with T2-KD cells and gRNA2 cell line being sensitive, while the gRNA1 cell line was resistant to PTX treatment. In addition, significant differences in the growth of gRNA1 and gRNA2 cell lines were observed in in vitro 3D cultures as well as in an in vivo mouse xenograft model. CONCLUSIONS Our results suggest that the inhibition of TIMP-2 by siRNA and CRISPR/Cas-9 modulate the expression of MMP-2 and MMP-14 and reprogram ovarian cancer cells to facilitate proliferation and invasion. Distinct disparities in in vitro chemosensitivity and growth in 3D culture, and differences in tumour burden and invasion to proximal organs in a mouse model imply that selective suppression of TIMP-2 expression by siRNA or CRISPR/Cas-9 alters important aspects of metastasis and chemosensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Ruth M. Escalona
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia ,Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - Simon Chu
- grid.1002.30000 0004 1936 7857Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia
| | - Elif Kadife
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - Jason K. Kelly
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia ,grid.1040.50000 0001 1091 4859School of Science, Psychology and Sport, Federation University, Mt Helen, VIC 3350 Australia
| | - Jock K. Findlay
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia
| | - Nuzhat Ahmed
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia ,Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia ,grid.1040.50000 0001 1091 4859School of Science, Psychology and Sport, Federation University, Mt Helen, VIC 3350 Australia
| |
Collapse
|
43
|
Kim JG, Kim SI, Song SH, Gu JY, Lee M, Kim HK. Diagnostic and prognostic role of circulating neutrophil extracellular trap markers and prekallikrein in patients with high-grade serous ovarian cancer. Front Oncol 2022; 12:992056. [PMID: 36620601 PMCID: PMC9813379 DOI: 10.3389/fonc.2022.992056] [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: 07/12/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Tumor-promoting inflammation is among the hallmarks of cancer. Prekallikrein is among the acute-phase reactants in the inflammatory response; moreover, neutrophils release nuclear contents into the extracellular space to create neutrophil extracellular traps (NET). We aimed to investigate the diagnostic and prognostic utilities of circulating plasma NET markers and prekallikrein for high-grade serous ovarian cancer (HGSOC). Methods Circulating levels of three NET markers (histone-DNA complex, cell-free DNA, and neutrophil elastase) and prekallikrein were measured in 75 patients with HGSOC and 23 healthy controls. We used an area under the receiver operating characteristic curve (AUC) analysis to investigate their diagnostic and prognostic utilities for HGSOC. Results Compared with healthy controls, patients with HGSOC showed significantly higher levels of the three NET markers and prekallikrein. Patients with advanced-stage HGSOC showed significantly higher levels of the cell-free DNA (87.4 vs. 79.5 ng/ml; P = 0.013), compared with those with early-stage HGSOC. Further, the levels of histone-DNA complex, neutrophil elastase, and prekallikrein did not significantly differ according to the cancer stage. All markers showed significant diagnostic utility. Notably, a logistic regression-based model that comprised all four markers showed the strongest diagnostic power (AUC, 0.966; 95% confidence interval [CI], 0.933-1.000). Specifically, neutrophil elastase was identified as an independent poor prognostic factor for overall survival (adjusted hazard ratio [aHR], 10.17; 95% CI, 1.09-94.97; P = 0.042) and progression-free survival (aHR, 14.47; 95% CI, 1.52-137.35; P = 0.020) in patients with HGSOC. Conclusions The levels of the three NET markers and prekallikrein might be novel diagnostic and prognostic markers for HGSOC.
Collapse
Affiliation(s)
- Jisoo G. Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Ja-Yoon Gu
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea,Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea,*Correspondence: Maria Lee, ; Hyun Kyung Kim,
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea,*Correspondence: Maria Lee, ; Hyun Kyung Kim,
| |
Collapse
|
44
|
Clemente-González C, Carnero A. Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation. Cancers (Basel) 2022; 14:5930. [PMID: 36497411 PMCID: PMC9738438 DOI: 10.3390/cancers14235930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
During tumor growth, the delivery of oxygen to cells is impaired due to aberrant or absent vasculature. This causes an adaptative response that activates the expression of genes that control several essential processes, such as glycolysis, neovascularization, immune suppression, and the cancer stemness phenotype, leading to increased metastasis and resistance to therapy. Hypoxic tumor cells also respond to an altered hypoxic microenvironment by secreting vesicles, factors, cytokines and nucleic acids that modify not only the immediate microenvironment but also organs at distant sites, allowing or facilitating the attachment and growth of tumor cells and contributing to metastasis. Hypoxia induces the release of molecules of different biochemical natures, either secreted or inside extracellular vesicles, and both tumor cells and stromal cells are involved in this process. The mechanisms by which these signals that can modify the premetastatic niche are sent from the primary tumor site include changes in the extracellular matrix, recruitment and activation of different stromal cells and immune or nonimmune cells, metabolic reprogramming, and molecular signaling network rewiring. In this review, we will discuss how hypoxia might alter the premetastatic niche through different signaling molecules.
Collapse
Affiliation(s)
- Cynthia Clemente-González
- Instituto de Biomedicina de Sevilla (IBIS), Consejo Superior de Investigaciones Científicas, Hospital Universitario Virgen del Rocío (HUVR), Universidad de Sevilla, 41013 Seville, Spain
- CIBERONC (Centro de Investigación Biomédica en Red Cáncer), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Consejo Superior de Investigaciones Científicas, Hospital Universitario Virgen del Rocío (HUVR), Universidad de Sevilla, 41013 Seville, Spain
- CIBERONC (Centro de Investigación Biomédica en Red Cáncer), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
45
|
Rašková M, Lacina L, Kejík Z, Venhauerová A, Skaličková M, Kolář M, Jakubek M, Rosel D, Smetana K, Brábek J. The Role of IL-6 in Cancer Cell Invasiveness and Metastasis-Overview and Therapeutic Opportunities. Cells 2022; 11:3698. [PMID: 36429126 PMCID: PMC9688109 DOI: 10.3390/cells11223698] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Interleukin 6 (IL-6) belongs to a broad class of cytokines involved in the regulation of various homeostatic and pathological processes. These activities range from regulating embryonic development, wound healing and ageing, inflammation, and immunity, including COVID-19. In this review, we summarise the role of IL-6 signalling pathways in cancer biology, with particular emphasis on cancer cell invasiveness and metastasis formation. Targeting principal components of IL-6 signalling (e.g., IL-6Rs, gp130, STAT3, NF-κB) is an intensively studied approach in preclinical cancer research. It is of significant translational potential; numerous studies strongly imply the remarkable potential of IL-6 signalling inhibitors, especially in metastasis suppression.
Collapse
Affiliation(s)
- Magdalena Rašková
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Lukáš Lacina
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Zdeněk Kejík
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Anna Venhauerová
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Markéta Skaličková
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Kolář
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, 140 00 Prague, Czech Republic
| | - Milan Jakubek
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Karel Smetana
- Centre for Tumour Ecology, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| |
Collapse
|
46
|
Adaptive Auricular Point Acupressure for Sleep Disturbance in Women with Breast Cancer: A Randomized Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8637386. [PMID: 36353150 PMCID: PMC9640241 DOI: 10.1155/2022/8637386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Objectives The study aimed to evaluate the preliminary effect and efficacy of auricular point acupressure (APA) on the quality of sleep in women with breast cancer who were undergoing chemotherapy. Sample & Setting. We conducted a randomized controlled trial on 68 patients with breast cancer who reported poor sleep quality based on the Pittsburgh Sleep Quality Index (PSQI) scores (>7). Methods & Variables. Participants were randomly assigned to an APA treatment group or a control group. Patients in the APA group had magnetic pellets attached to selected auricular points and were instructed to apply pressure to these points 4×/day for three consecutive weeks. We objectively measured sleep quality using the Actiwatch Spectrum and the PSQI at the baseline and postintervention. Statistical analyses of changes in sleep data were performed using the t-test, a rank-sum test, and analyses of covariance. Results In patients treated with APA, the PSQI total score and sleep onset latency had significantly decreased, while the total sleep time and sleep efficiency had significantly increased. Although the total PSQI score differed between groups at the baseline, ANCOVA results showed that the APA group had a significantly lower total PSQI score. Conclusion APA could be an inexpensive and effective approach to improving sleep quality and reducing sleep disturbance in patients with breast cancer. Further research needs a larger sample size to verify our findings.
Collapse
|
47
|
Singla RK, Sharma P, Kumar D, Gautam RK, Goyal R, Tsagkaris C, Dubey AK, Bansal H, Sharma R, Shen B. The role of nanomaterials in enhancing natural product translational potential and modulating endoplasmic reticulum stress in the treatment of ovarian cancer. Front Pharmacol 2022; 13:987088. [PMID: 36386196 PMCID: PMC9643842 DOI: 10.3389/fphar.2022.987088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/03/2022] [Indexed: 10/21/2023] Open
Abstract
Ovarian cancer, and particularly its most frequent type, epithelial ovarian carcinoma, constitutes one of the most dangerous malignant tumors among females. Substantial evidence has described the potential of phytochemicals against ovarian cancer. The effect of natural compounds on endoplasmic reticulum (ER) stress is of great relevance in this regard. In ovarian cancer, the accumulation of misfolded proteins in the ER lumen results in decompensated ER stress. This leads to deregulation in the physiological processes for the posttranslational modification of proteins, jeopardizes cellular homeostasis, and increases apoptotic signaling. Several metabolites and metabolite extracts of phytochemical origin have been studied in the context of ER stress in ovarian cancer. Resveratrol, quercetin, curcumin, fucosterol, cleistopholine, fucoidan, and epicatechin gallate, among others, have shown inhibitory potential against ER stress. The chemical structure of each compound plays an important role concerning its pharmacodynamics, pharmacokinetics, and overall effectiveness. Studying and cross-comparing the chemical features that render different phytochemicals effective in eliciting particular anti-ER stress actions can help improve drug design or develop multipotent combination regimens. Many studies have also investigated the properties of formulations such as nanoparticles, niosomes, liposomes, and intravenous hydrogel based on curcumin and quercetin along with some other phytomolecules in ovarian cancer. Overall, the potential of phytochemicals in targeting genetic mechanisms of ovarian cancer warrants further translational and clinical investigation.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Pooja Sharma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
- Khalsa College of Pharmacy, Amritsar, India
| | - Dinesh Kumar
- Chitkara University School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Rupesh K. Gautam
- Department of Pharmacology, Indore Institute of Pharmacy, IIST Campus, Opposite IIM Indore, Indore, India
| | - Rajat Goyal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | | | | | - Himangini Bansal
- Delhi Institute of Pharmaceutical Sciences and Research, New Delhi, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Main C, Chen X, Zhao M, Chamley LW, Chen Q. Understanding How Pregnancy Protects Against Ovarian and Endometrial Cancer Development: Fetal Antigens May Be Involved. Endocrinology 2022; 163:6675223. [PMID: 36004540 PMCID: PMC9574549 DOI: 10.1210/endocr/bqac141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Indexed: 11/19/2022]
Abstract
It is well known that many factors, including infertility, obesity, type 2 diabetes, and family history of cancer, increase the risk of developing endometrial and ovarian cancer. However, multiparous women are known to have a lower risk of developing either ovarian or endometrial cancer than nonparous women. The lack of ovulation and shifting of sex hormonal balance, with decreased estrogen levels and increased progesterone levels during pregnancy, has traditionally been thought to be the major contributor to this decreased risk. However, in reality, the mechanisms underlying this phenomenon are relatively unknown. Increasing evidence suggests that endocrine factors are unlikely to completely explain the protective effect of pregnancies, and that multiple other nonendocrine mechanisms including fetal antigens and the newly proposed dormant cells hypothesis may also be involved. In this review, we summarize recent evidence and describe the potential underlying mechanisms that may explain how pregnancy protects against the development of ovarian and endometrial cancers in women's later life.
Collapse
Affiliation(s)
- Claudia Main
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Xinyue Chen
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Min Zhao
- Department of Gynecological Cancer, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 214002, China
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland 1141, New Zealand
| | - Qi Chen
- Correspondence: Qi Chen, MD, PhD, Department of Obstetrics and Gynaecology, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand. ; or Min Zhao, MD, PhD, Department of Gynecological Cancer, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, China.
| |
Collapse
|
49
|
Kuźmycz O, Kowalczyk A, Stączek P. Biological Activity of fac-[Re(CO) 3(phen)(aspirin)], fac-[Re(CO) 3(phen)(indomethacin)] and Their Original Counterparts against Ishikawa and HEC-1A Endometrial Cancer Cells. Int J Mol Sci 2022; 23:ijms231911568. [PMID: 36232870 PMCID: PMC9569891 DOI: 10.3390/ijms231911568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are inhibitors of cyclooxygenase enzyme (COX) and were found to have positive effects in reducing the risk of developing gynecological cancers. However, long-term administration of NSAIDs carries the risk of various side effects, including those in the digestive and circulatory systems. Therefore, there is a constant need to develop new NSAID derivatives. In this work, we investigated rhenium NSAIDs, comparing their effects on endometrial cancer cells with original NSAIDs, demonstrating the high activity of aspirin and indomethacin derivatives. The cytotoxic activity of rhenium derivatives against the Ishikawa and HEC-1A cancer cell lines was higher than that of the original NSAIDs. The IC50 after 24-h incubation of Ishikawa and HEC-1A were 188.06 µM and 394.06 µM for rhenium aspirin and 228.6 µM and 1459.3 µM for rhenium indomethacin, respectively. At the same time, IC50 of aspirin and indomethacin were 10,024.42 µM and 3295.3 µM for Ishikawa, and 27,255.8 µM and 5489.3 µM for HEC-1A, respectively. Moreover, these derivatives were found to inhibit the proliferation of both cell lines in a time- and state-dependent manner. The Ishikawa cell proliferation was strongly inhibited by rhenium aspirin and rhenium indomethacin after 72-h incubation (*** = p < 0.001), while the HEC-1A proliferation was inhibited by the same agents already after 24-h incubation (*** = p < 0.001). Furthermore, the ROS level in the mitochondria of the tested cells generated in the presence of rhenium derivatives was higher than the original NSAIDs. That was associated with rhenium indomethacin exclusively, which had a significant effect (*** = p < 0.001) on both Ishikawa and HEC-1A cancer cells. Rhenium aspirin had a significant effect (*** = p < 0.001) on the mitochondrial ROS level of Ishikawa cells only. Overall, the research revealed a high potential of the rhenium derivatives of aspirin and indomethacin against endometrial cancer cells compared with the original NSAIDs.
Collapse
|
50
|
Sánchez-Prieto M, Sánchez-Borrego R, Lubián-López DM, Pérez-López FR. Etiopathogenesis of ovarian cancer. An inflamm-aging entity? Gynecol Oncol Rep 2022; 42:101018. [PMID: 35719320 PMCID: PMC9198811 DOI: 10.1016/j.gore.2022.101018] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Ovarian cancer (OvCa) is a multifactorial disease. Several factors are involved in age-related increases in carcinogenesis. Exposure to inflammatory mediators contributes to increased cell division and genetic and epigenetic changes. We discuss the current carcinogenic hypotheses, sites of origin, and etiological factors of OvCa.
Ovarian cancer is one of the most common gynecologic cancers and has the highest mortality rate. The risk/protective factors of ovarian cancer suggest that its etiology is multifactorial. Several factors are involved in age-related increases in carcinogenesis, including the accumulation of senescent cells, inflammaging (a chronic inflammatory state that persists in the elderly), and immunosenescence (aging of the immune system) changes associated with poor immune surveillance. At sites of inflammation, exposure to high levels of inflammatory mediators, such as reactive oxygen species, cytokines, prostaglandins, and growth factors, contributes to increased cell division and genetic and epigenetic changes. These exposure-induced changes promote excessive cell proliferation, increased survival, malignant transformation, and cancer development. Furthermore, the proinflammatory tumor microenvironment contributes to ovarian cancer metastasis and chemoresistance. This narrative review of the literature was carried out to delineate the possible role of inflammaging in the etiopathogenesis of ovarian cancer development. We discuss the current carcinogenic hypotheses, sites of origin, and etiological factors of ovarian cancer. Treatment of inflammation may represent an attractive strategy for both the prevention and therapy of ovarian cancer.
Collapse
Affiliation(s)
- Manuel Sánchez-Prieto
- Department of Obstetrics and Gynecology, Instituto Universitario Dexeus, Barcelona, Spain
- Corresponding author at: Instituto Universitario Dexeus, Sabino de Arana 5-19, 08028 Barcelona, Spain.
| | | | | | | |
Collapse
|