1
|
Doran BR, Moffitt LR, Wilson AL, Stephens AN, Bilandzic M. Leader Cells: Invade and Evade-The Frontline of Cancer Progression. Int J Mol Sci 2024; 25:10554. [PMID: 39408880 PMCID: PMC11476628 DOI: 10.3390/ijms251910554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Metastasis is the leading cause of cancer-related mortality; however, a complete understanding of the molecular programs driving the metastatic cascade is lacking. Metastasis is dependent on collective invasion-a developmental process exploited by many epithelial cancers to establish secondary tumours and promote widespread disease. The key drivers of collective invasion are "Leader Cells", a functionally distinct subpopulation of cells that direct migration, cellular contractility, and lead trailing or follower cells. While a significant body of research has focused on leader cell biology in the traditional context of collective invasion, the influence of metastasis-promoting leader cells is an emerging area of study. This review provides insights into the expanded role of leader cells, detailing emerging evidence on the hybrid epithelial-mesenchymal transition (EMT) state and the phenotypical plasticity exhibited by leader cells. Additionally, we explore the role of leader cells in chemotherapeutic resistance and immune evasion, highlighting their potential as effective and diverse targets for novel cancer therapies.
Collapse
Affiliation(s)
- Brittany R. Doran
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Laura R. Moffitt
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| |
Collapse
|
2
|
Chen B, Liu J. Advances in ovarian tumor stem cells and therapy. Cell Biochem Biophys 2024; 82:1871-1892. [PMID: 38955927 DOI: 10.1007/s12013-024-01385-8] [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] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer is considered the most lethal among all gynecological malignancies due to its early metastatic dissemination, extensive spread, and malignant ascites. The current standard of care for advanced ovarian cancer involves a combination of cytoreductive surgery and chemotherapy utilizing platinum-based and taxane-based agents. Although initial treatment yields clinical remission in 70-80% of patients, the majority eventually develop treatment resistance and tumor recurrence. A growing body of evidence indicates the existence of cancer stem cells within diverse solid tumors, including ovarian cancer, which function as a subpopulation to propel tumor growth and disease advancement by means of drug resistance, recurrence, and metastasis. The presence of ovarian cancer stem cells is widely considered to be a significant contributor to the unfavorable clinical outcomes observed in patients with ovarian cancer, as they play a crucial role in mediating chemotherapy resistance, recurrence, and metastasis. Ovarian cancer stem cells possess the capacity to reassemble within the entirety of the tumor following conventional treatment, thereby instigating the recurrence of ovarian cancer and inducing resistance to treatment. Consequently, the creation of therapeutic approaches aimed at eliminating ovarian cancer stem cells holds great potential for the management of ovarian cancer. These cells are regarded as one of the most auspicious targets and mechanisms for the treatment of ovarian cancer. There is a pressing need for a comprehensive comprehension of the fundamental mechanisms of ovarian cancer's recurrence, metastasis, and drug resistance, alongside the development of effective strategies to overcome chemoresistance, metastasis, and recurrence. The implementation of cancer stem cell therapies may potentially augment the tumor cells' sensitivity to existing chemotherapy protocols, thereby mitigating the risks of tumor metastasis and recurrence, and ultimately improving the survival rates of ovarian cancer patients.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | - Jiaqi Liu
- Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
3
|
Zhang M, Yin R, Li K. Advances and challenges in the origin and evolution of ovarian cancer organoids. Front Oncol 2024; 14:1429141. [PMID: 39220646 PMCID: PMC11362079 DOI: 10.3389/fonc.2024.1429141] [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: 05/07/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Despite advancements in cancer research, epithelial ovarian cancer remains a leading threat to women's health with a low five-year survival rate of 48%. Prognosis for advanced cases, especially International Federation of Gynecology and Obstetrics (FIGO) III-IV, is poor. Standard care includes surgical resection and platinum-based chemo, but 70-80% face recurrence and chemoresistance. In recent years, three- dimensional (3D) cancer models, especially patients-derived organoids (PDOs), have revolutionized cancer research for personalized treatment. By transcending the constraints of conventional models, organoids accurately recapitulate crucial morphological, histological, and genetic characteristics of diseases, particularly in the context of ovarian cancer. The extensive potential of ovarian cancer organoids is explored, spanning from foundational theories to cutting-edge applications. As potent preclinical models, organoids offer invaluable tools for predicting patient treatment responses and guiding the development of personalized therapeutic strategies. Furthermore, in the arena of drug evaluation, organoids demonstrate their unique versatility as platforms, enabling comprehensive testing of innovative drug combinations and novel candidates, thereby pioneering new avenues in pharmaceutical research. Notably, organoids mimic the dynamic progression of ovarian cancer, from inception to systemic dissemination, shedding light on intricate and subtle disease mechanisms, and providing crucial insights. Operating at an individualized level, organoids also unravel the complex mechanisms underlying drug resistance, presenting strategic opportunities for the development of effective treatment strategies. This review summarizes the emerging role of ovarian cancer organoids, meticulously cultivated cellular clusters within three-dimensional models, as a groundbreaking paradigm in research.
Collapse
Affiliation(s)
- Mengpei Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Rutie Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Kemin Li
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
4
|
Pan Y, Yang X, Chen M, Shi K, Lyu Y, Meeson AP, Lash GE. Role of Cancer Side Population Stem Cells in Ovarian Cancer Angiogenesis. Med Princ Pract 2024; 33:403-413. [PMID: 39068919 PMCID: PMC11460956 DOI: 10.1159/000539642] [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: 01/05/2024] [Accepted: 06/03/2024] [Indexed: 07/30/2024] Open
Abstract
Ovarian cancer is one of the most common gynecologic malignancies. Recurrence and metastasis often occur after treatment, and it has the highest mortality rate of all gynecological tumors. Cancer stem cells (CSCs) are a small population of cells with the ability of self-renewal, multidirectional differentiation, and infinite proliferation. They have been shown to play an important role in tumor growth, metastasis, drug resistance, and angiogenesis. Ovarian cancer side population (SP) cells, a type of CSC, have been shown to play roles in tumor formation, colony formation, xenograft tumor formation, ascites formation, and tumor metastasis. The rapid progression of tumor angiogenesis is necessary for tumor growth; however, many of the mechanisms driving this process are unclear as is the contribution of CSCs. The aim of this review was to document the current state of knowledge of the molecular mechanism of ovarian cancer stem cells (OCSCs) in regulating tumor angiogenesis.
Collapse
Affiliation(s)
- Yue Pan
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - XueFen Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Miaojuan Chen
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kun Shi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yuan Lyu
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | | | - Gendie E. Lash
- Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Third Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Sun J, Feng Q, He Y, Wang M, Wu Y. Lactate activates CCL18 expression via H3K18 lactylation in macrophages to promote tumorigenesis of ovarian cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1373-1386. [PMID: 39010846 PMCID: PMC11543520 DOI: 10.3724/abbs.2024111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/02/2024] [Indexed: 07/17/2024] Open
Abstract
This study investigates the role of lactate in the genesis and progression of ovarian cancer (OV) and explores the underlying mechanisms. Serum lactate levels show a positive correlation with tumor grade and poor prognosis in patients with OV. Bioinformatics analysis identifies CCL18 as a lactate-related gene in OV. CCL18 is up-regulated in cancerous tissues and positively related to serum lactate levels in OV patients. THP-1 cells are exposed to phorbol-12-myristate-13-acetate for M0 macrophage induction. The results of RT-qPCR and ELISA for M1/M2 macrophage-related markers and inflammatory cytokines show that the exposure of lactate to macrophages induces M2 polarization. Based on the coculture of OV cells with macrophages, lactate-treated macrophages induces a significant increase in the proliferation and migration of OV cells. However, these effects can be reversed by silencing of Gpr132 in macrophages or treatment with anti-CCL18 antibody. Experiments using the xenograft model verify that the oncogenic role of lactate in tumor growth and metastasis relies on Gpr132 and CCL18. ChIP-qPCR and luciferase reporter assays reveal that lactate regulates CCL18 expression via H3K18 lactylation. In conclusion, lactate is a potential therapeutic target for OV. It is involved in tumorigenesis by activating CCL18 expression via H3K18 lactylation in macrophages.
Collapse
Affiliation(s)
- Jinrui Sun
- Department of GynecologyShanxi Provincial People’s HospitalTaiyuan030001China
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Qinmei Feng
- Department of GynecologyShanxi Provincial People’s HospitalTaiyuan030001China
| | - Yue He
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Ming Wang
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Yumei Wu
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| |
Collapse
|
6
|
Zhao X, Yang P, Liu L, Li Y, Huang Y, Tang H, Zhou Y, Mao Y. Optimal debulking surgery in ovarian cancer patients: MRI may predict the necessity of rectosigmoid resection. Insights Imaging 2024; 15:145. [PMID: 38886313 PMCID: PMC11183003 DOI: 10.1186/s13244-024-01725-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVES To determine whether MRI can predict the necessity of rectosigmoid resection (RR) for optimal debulking surgery (ODS) in ovarian cancer (OC) patients and to compare the predictive accuracy of pre- and post-neoadjuvant chemotherapy (NACT) MRI. METHODS The MRI of 82 OC were retrospectively analyzed, including six bowel signs (length, transverse axis, thickness, circumference, muscularis involvement, and submucosal edema) and four para-intestinal signs (vaginal, parametrial, ureteral, and sacro-recto-genital septum involvement). The parameters reflecting the degree of muscularis involvement were measured. Patients were divided into non-RR and RR groups based on the operation and postoperative outcomes. The independent predictors of the need for RR were identified by multivariate logistic regression analysis. RESULTS Imaging for 82 patients was evaluated (67 without and 15 with NACT). Submucosal edema and muscularis involvement (OR 13.33 and 8.40, respectively) were independent predictors of the need for RR, with sensitivities of 83.3% and 94.4% and specificities of 93.9% and 81.6%, respectively. Among the parameters reflecting the degree of muscularis involvement, circumference ≥ 3/12 had the highest prediction accuracy, increasing the specificity from 81.6% for muscularis involvement only to 98.0%, with only a slight decrease in sensitivity (from 94.4% to 88.9%). The predictive sensitivities of pre-NACT and post-NACT MRI were 100.0% and 12.5%, respectively, and the specificities were 85.7% and 100.0%, respectively. CONCLUSIONS MRI analysis of rectosigmoid muscularis involvement and its circumference can help predict the necessity of RR in OC patients, and pre-NACT MRI may be more suitable for evaluation. CRITICAL RELEVANCE STATEMENT We analyzed preoperative pelvic MRI in OC patients. Our findings suggest that MRI has predictive potential for identifying patients who require RR to achieve ODS. KEY POINTS The need for RR must be determined to optimize treatment for OC patients. Muscularis involvement circumference ≥ 3/12 could help predict RR. Pre-NACT MRI may be superior to post-NACT MRI in predicting RR.
Collapse
Affiliation(s)
- Xiaofang Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Yang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Huang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huali Tang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Zhou
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yun Mao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Battistini C, Kenny HA, Zambuto M, Nieddu V, Melocchi V, Decio A, Lo Riso P, Villa CE, Gatto A, Ghioni M, Porta FM, Testa G, Giavazzi R, Colombo N, Bianchi F, Lengyel E, Cavallaro U. Tumor microenvironment-induced FOXM1 regulates ovarian cancer stemness. Cell Death Dis 2024; 15:370. [PMID: 38806454 PMCID: PMC11133450 DOI: 10.1038/s41419-024-06767-7] [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: 04/09/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
In ovarian tumors, the omental microenvironment profoundly influences the behavior of cancer cells and sustains the acquisition of stem-like traits, with major impacts on tumor aggressiveness and relapse. Here, we leverage a patient-derived platform of organotypic cultures to study the crosstalk between the tumor microenvironment and ovarian cancer stem cells. We discovered that the pro-tumorigenic transcription factor FOXM1 is specifically induced by the microenvironment in ovarian cancer stem cells, through activation of FAK/YAP signaling. The microenvironment-induced FOXM1 sustains stemness, and its inactivation reduces cancer stem cells survival in the omental niche and enhances their response to the PARP inhibitor Olaparib. By unveiling the novel role of FOXM1 in ovarian cancer stemness, our findings highlight patient-derived organotypic co-cultures as a powerful tool to capture clinically relevant mechanisms of the microenvironment/cancer stem cells crosstalk, contributing to the identification of tumor vulnerabilities.
Collapse
Affiliation(s)
- Chiara Battistini
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, 20139, Milan, Italy
| | - Hilary A Kenny
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL, 60637, USA
| | - Melissa Zambuto
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, 20139, Milan, Italy
| | - Valentina Nieddu
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, 20139, Milan, Italy
| | - Valentina Melocchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, FG, Italy
| | - Alessandra Decio
- Laboratory of Cancer Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, 20156, Milan, Italy
| | - Pietro Lo Riso
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
| | | | - Alessia Gatto
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, 20139, Milan, Italy
| | - Mariacristina Ghioni
- Division of Pathology, European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Francesca M Porta
- Division of Pathology, European Institute of Oncology IRCCS, 20141, Milan, Italy
- School of Pathology, University of Milan, 20122, Milan, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Cancer Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, 20156, Milan, Italy
| | - Nicoletta Colombo
- Division of Gynecologic Oncology, European Institute of Oncology IRCCS, 20141, Milan, Italy
- University of Milan-Bicocca, 20126, Milan, Italy
| | - Fabrizio Bianchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, FG, Italy
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL, 60637, USA
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, 20139, Milan, Italy.
| |
Collapse
|
8
|
Kwon JE, Jang Y, Yun BS, Kang S, Kim YH, Kim BG, Cho NH. MET overexpression in ovarian cancer via CD24-induced downregulation of miR-181a: A signalling for cellular quiescence-like state and chemoresistance in ovarian CSCs. Cell Prolif 2024; 57:e13582. [PMID: 38030594 PMCID: PMC11056702 DOI: 10.1111/cpr.13582] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Increased expression of CD24 and MET, markers for cancer stem-like cells (CSCs), are each associated with ovarian cancer severity. However, whether CD24 and MET are co-expressed in ovarian CSCs and, if so, how they are related to CSC phenotype manifestation remains unknown. Our immunohistochemistry analysis showed that the co-expression of CD24 and MET was associated with poorer patient survival in ovarian cancer than those without. In addition, analyses using KM plotter and ROC plotter presented that the overexpression of CD24 or MET in ovarian cancer patients was associated with resistance to platinum-based chemotherapy. In our miRNA transcriptome and putative target genes analyses, miR-181a was downregulated in CD24-high ovarian cancer cells compared to CD24-low and predicted to bind to CD24 and MET 3'UTRs. In OV90 and SK-OV-3 cells, CD24 downregulated miR-181a expression by Src-mediated YY1 activation, leading to increased expression of MET. And, CD24 or MET knockdown or miR-181a overexpression inhibited the manifestation of CSC phenotypes, cellular quiescence-like state and chemoresistance, in OV90 and SK-OV-3 cells: increased colony formation, decreased G0/G1 phase cell population and increased sensitivity to Cisplatin and Carboplatin. Our findings suggest that CD24-miR-181a-MET may consist of a signalling route for ovarian CSCs, therefore being a combinatory set of markers and therapeutic targets for ovarian CSCs.
Collapse
Affiliation(s)
- Ji Eun Kwon
- Department of PathologyAjou University School of MedicineSuwonKorea
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoulKorea
| | - Yeonsue Jang
- Department of PathologyYonsei University College of MedicineSeoulKorea
| | - Bo Seong Yun
- Department of Gynecology Obstetrics and Gynecology, CHA Gangnam Medical CenterCHA UniversitySeoulKorea
| | - Suki Kang
- Department of PathologyYonsei University College of MedicineSeoulKorea
| | - Yon Hee Kim
- Department of PathologySoonchunhyang University HospitalSeoulKorea
| | - Baek Gil Kim
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoulKorea
- Department of PathologyYonsei University College of MedicineSeoulKorea
| | - Nam Hoon Cho
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoulKorea
- Department of PathologyYonsei University College of MedicineSeoulKorea
- Severance Biomedical Science Institute (SBSI)Yonsei University College of MedicineSeoulKorea
| |
Collapse
|
9
|
Islam SS, Al-Mohanna FH, Yousef IM, Al-Badawi IA, Aboussekhra A. Ovarian tumor cell-derived JAGGED2 promotes omental metastasis through stimulating the Notch signaling pathway in the mesothelial cells. Cell Death Dis 2024; 15:247. [PMID: 38575576 PMCID: PMC10995149 DOI: 10.1038/s41419-024-06512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 04/06/2024]
Abstract
The primary site of metastasis for epithelial ovarian cancer (EOC) is the peritoneum, and it occurs through a multistep process that begins with adhesive contacts between cancer cells and mesothelial cells. Despite evidence that Notch signaling has a role in ovarian cancer, it is unclear how exactly it contributes to ovarian cancer omental metastasis, as well as the cellular dynamics and intrinsic pathways that drive this tropism. Here we show that tumor cells produced the Notch ligand Jagged2 is a clinically and functionally critical mediator of ovarian cancer omental metastasis by activating the Notch signaling in single-layered omental mesothelial cells. In turn, Jagged2 promotes tumor growth and therapeutic resistance by stimulating IL-6 release from mesothelial cells. Additionally, Jagged2 is a potent downstream mediator of the omental metastasis cytokine TGF-β that is released during omental destruction. Importantly, therapeutic inhibition of Jagged2-mediated omental metastasis was significantly improved by directly disrupting the Notch pathway in omental mesothelial cells. These findings highlight the key role of Jagged2 to the functional interplay between the TGF-β and the Notch signaling pathways during the metastatic process of ovarian cancer cells to the omentum and identify the Notch signaling molecule as a precision therapeutic target for ovarian cancer metastasis.
Collapse
Affiliation(s)
- Syed S Islam
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.
- School of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| | - Falah H Al-Mohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Iman M Yousef
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ismail A Al-Badawi
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.
| |
Collapse
|
10
|
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
|
11
|
El Baba R, Haidar Ahmad S, Monnien F, Mansar R, Bibeau F, Herbein G. Polyploidy, EZH2 upregulation, and transformation in cytomegalovirus-infected human ovarian epithelial cells. Oncogene 2023; 42:3047-3061. [PMID: 37634008 PMCID: PMC10555822 DOI: 10.1038/s41388-023-02813-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/28/2023]
Abstract
Human cytomegalovirus (HCMV) infection has been implicated in epithelial ovarian cancer (OC). Polyploidy giant cancer cells (PGCCs) have been observed in high-grade serous ovarian carcinoma (HGSOC); they possess cancer stem cell-like characteristics and give rise to progeny cells expressing epithelial-mesenchymal transition (EMT) markers. EZH2 plays a potential oncogenic role, correlating with high proliferative index and tumor grade in OC. Herein, we present the experimental evidence for HCMV as a reprogramming vector that elicited human ovarian epithelial cells (OECs) transformation leading to the generation of "CMV-transformed Ovarian cells" (CTO). The infection with the two high-risk clinical strains, namely HCMV-DB and BL provoked a distinct cellular and molecular mechanisms in infected OECs. EZH2 upregulation and cellular proliferation were curtailed by using EZH2 inhibitors. The HGSOC biopsies were characterized by an elevated EZH2 expression, possessing a strong positive correlation between the aforementioned marker and HCMV. From HGSOC biopsies, we isolated three HCMV clinical strains that transformed OECs generating CTO cells which displayed proliferative potentials in addition to EZH2 upregulation and PGCCs generation; these features were reduced upon EZH2 inhibition. High-risk HCMV strains transformed OECs confirming an HCMV-induced epithelial ovarian cancer model and highlighting EZH2 tumorigenic properties. Our findings might be highly relevant in the pathophysiology of ovarian tumors thereby nominating new targeted therapeutics.
Collapse
Affiliation(s)
- Ranim El Baba
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | - Sandy Haidar Ahmad
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | | | - Racha Mansar
- Department of Pathology, CHU Besançon, Besançon, France
| | | | - Georges Herbein
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France.
- Department of Virology, CHU Besançon, Besançon, France.
| |
Collapse
|
12
|
Iżycka N, Zaborowski MP, Ciecierski Ł, Jaz K, Szubert S, Miedziarek C, Rezler M, Piątek-Bajan K, Synakiewicz A, Jankowska A, Figlerowicz M, Sterzyńska K, Nowak-Markwitz E. Cancer Stem Cell Markers-Clinical Relevance and Prognostic Value in High-Grade Serous Ovarian Cancer (HGSOC) Based on The Cancer Genome Atlas Analysis. Int J Mol Sci 2023; 24:12746. [PMID: 37628927 PMCID: PMC10454196 DOI: 10.3390/ijms241612746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer stem cells (CSCs) may contribute to an increased risk of recurrence in ovarian cancer (OC). Further research is needed to identify associations between CSC markers and OC patients' clinical outcomes with greater certainty. If they prove to be correct, in the future, the CSC markers can be used to help predict survival and indicate new therapeutic targets. This study aimed to determine the CSC markers at mRNA and protein levels and their association with clinical presentation, outcome, and risk of recurrence in HGSOC (High-Grade Serous Ovarian Cancer). TCGA (The Cancer Genome Atlas) database with 558 ovarian cancer tumor samples was used for the evaluation of 13 CSC markers (ALDH1A1, CD44, EPCAM, KIT, LGR5, NES, NOTCH3, POU5F1, PROM1, PTTG1, ROR1, SOX9, and THY1). Data on mRNA and protein levels assessed by microarray and mass spectrometry were retrieved from TCGA. Models to predict chemotherapy response and survival were built using multiple variables, including epidemiological data, expression levels, and machine learning methodology. ALDH1A1 and LGR5 mRNA expressions indicated a higher platinum sensitivity (p = 3.50 × 10-3; p = 0.01, respectively). POU5F1 mRNA expression marked platinum-resistant tumors (p = 9.43 × 10-3). CD44 and EPCAM mRNA expression correlated with longer overall survival (OS) (p = 0.043; p = 0.039, respectively). THY1 mRNA and protein levels were associated with worse OS (p = 0.019; p = 0.015, respectively). Disease-free survival (DFS) was positively affected by EPCAM (p = 0.004), LGR5 (p = 0.018), and CD44 (p = 0.012). In the multivariate model based on CSC marker expression, the high-risk group had 9.1 months longer median overall survival than the low-risk group (p < 0.001). ALDH1A1, CD44, EPCAM, LGR5, POU5F1, and THY1 levels in OC may be used as prognostic factors for the primary outcome and help predict the treatment response.
Collapse
Affiliation(s)
- Natalia Iżycka
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Mikołaj Piotr Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
- European Center for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland (M.F.)
| | - Łukasz Ciecierski
- European Center for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland (M.F.)
| | - Kamila Jaz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Sebastian Szubert
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Cezary Miedziarek
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Marta Rezler
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Kinga Piątek-Bajan
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Aneta Synakiewicz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| | - Anna Jankowska
- Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D St., 60-806 Poznan, Poland;
| | - Marek Figlerowicz
- European Center for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland (M.F.)
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 St., 61-781 Poznan, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznan, Poland (S.S.)
| |
Collapse
|
13
|
Bhat BA, Saifi I, Khamjan NA, Hamdani SS, Algaissi A, Rashid S, Alshehri MM, Ganie SA, Lohani M, Abdelwahab SI, Dar SA. Exploring the tumor immune microenvironment in ovarian cancer: a way-out to the therapeutic roadmap. Expert Opin Ther Targets 2023; 27:841-860. [PMID: 37712621 DOI: 10.1080/14728222.2023.2259096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/21/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Despite cancer treatment strides, mortality due to ovarian cancer remains high globally. While immunotherapy has proven effective in treating cancers with low cure rates, it has limitations. Growing evidence suggests that both tumoral and non-tumoral components of the tumor immune microenvironment (TIME) play a significant role in cancer growth. Therefore, developing novel and focused therapy for ovarian cancer is critical. Studies indicate that TIME is involved in developing ovarian cancer, particularly genome-, transcriptome-, and proteome-wide studies. As a result, TIME may present a prospective therapeutic target for ovarian cancer patients. AREAS COVERED We examined several TIME-targeting medicines and the connection between TIME and ovarian cancer. The key protagonists and events in the TIME and therapeutic strategies that explicitly target these events in ovarian cancer are discussed. EXPERT OPINION We highlighted various targeted therapies against TIME in ovarian cancer, including anti-angiogenesis therapies and immune checkpoint inhibitors. While these therapies are in their infancy, they have shown promise in controlling ovarian cancer progression. The use of 'omics' technology is helping in better understanding of TIME in ovarian cancer and potentially identifying new therapeutic targets. TIME-targeted strategies could account for an additional treatment strategy when treating ovarian cancer.
Collapse
Affiliation(s)
- Basharat Ahmad Bhat
- Department of Bioresources, Amar Singh College Campus, Cluster University, Srinagar, India
| | - Ifra Saifi
- Department of Botany, Chaudhary Charan Singh University, Meerut India
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Syed Suhail Hamdani
- Department of Bioresources, Amar Singh College Campus, Cluster University, Srinagar, India
| | - Abdullah Algaissi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Medical Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Safeena Rashid
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, India
| | | | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Mohtashim Lohani
- Department of Emergency Medical Services, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | | | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
14
|
Englisz A, Smycz-Kubańska M, Mielczarek-Palacz A. Evaluation of the Potential Diagnostic Utility of the Determination of Selected Immunological and Molecular Parameters in Patients with Ovarian Cancer. Diagnostics (Basel) 2023; 13:diagnostics13101714. [PMID: 37238197 DOI: 10.3390/diagnostics13101714] [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: 03/10/2023] [Revised: 04/28/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Ovarian cancer is one of the most serious challenges in modern gynaecological oncology. Due to its non-specific symptoms and the lack of an effective screening procedure to detect the disease at an early stage, ovarian cancer is still marked by a high mortality rate among women. For this reason, a great deal of research is being carried out to find new markers that can be used in the detection of ovarian cancer to improve early diagnosis and survival rates of women with ovarian cancer. Our study focuses on presenting the currently used diagnostic markers and the latest selected immunological and molecular parameters being currently investigated for their potential use in the development of new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Aleksandra Englisz
- The Doctoral School, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marta Smycz-Kubańska
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
15
|
Yang L, Yang M, Cui C, Long X, Li Y, Dai W, Lang T, Zhou Q. The myo-inositol biosynthesis rate-limiting enzyme ISYNA1 suppresses the stemness of ovarian cancer via Notch1 pathway. Cell Signal 2023; 107:110688. [PMID: 37105506 DOI: 10.1016/j.cellsig.2023.110688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 04/29/2023]
Abstract
Cancer stem cells (CSCs) play a central role in ovarian cancer (OC), understanding regulatory mechanisms governing their stemness is critical. Here, we report ISYNA1, the rate-limiting enzyme in myo-inositol biosynthesis, as a suppressor of OC regulating cancer stemness. We identified ISYNA1 as a differentially expressed gene in normal ovary and ovarian cancer tissues, as well as OC cells and OCSCs. Low ISYNA1 expression correlated with poor prognosis in OC patients. In addition, ISYNA1 was negatively correlated with CSC markers, and ISYNA1-related pathways were enriched in Wnt, Notch, and other critical cancer pathways. ISYNA1 deficiency promoted OC cell growth, migration, and invasion ability in vitro and in vivo. Knockdown of ISYNA1 increased stemness of OC cells, including self-renewal, CSC markers expression, ALDH activity, and proportion of CD44+/CD117+ CSCs. Conversely, ectopic overexpression of ISYNA1 suppresses cell proliferation, migration, invasion and stemness of OC cells. Mechanistically, ISYNA1 inhibits OC stemness by regulating myo-inositol to suppress Notch1 signaling. In summary, these data provide evidence that ISYNA1 act as a tumor suppressor in OC and a regulator of stemness, providing insight into potentially targetable pathways for ovarian cancer therapy.
Collapse
Affiliation(s)
- Lingling Yang
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Muyao Yang
- College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China
| | - Chenxi Cui
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xingtao Long
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Yunzhe Li
- College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China
| | - Weili Dai
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China.
| | - Qi Zhou
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China; Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China.
| |
Collapse
|
16
|
Sohn EJ, Kim JH, Oh SO, Kim JY. Regulation of self-renewal in ovarian cancer stem cells by fructose via chaperone-mediated autophagy. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166723. [PMID: 37087023 DOI: 10.1016/j.bbadis.2023.166723] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
The chaperone-mediated autophagy (CMA) pathway is deregulated in different types of cancers; however, its role in cancer stem cells (CSCs) is unknown yet. Development of ovarian cancer, the most lethal gynecological type of cancer, involves the metastasis of CSCs to the abdominal cavity. This study aims to determine the role of CMA in ovarian CSCs. We found that the transcription factor EB (TFEB) and trehalose, a disaccharide that induces TFEB activation, enhance the expression of octamer-binding transcription factor 4 (OCT4) stem cell and lysosomal-associated membrane protein 2A (LAMP2A) CMA markers. However, trehalose did not increase the level of the LC3II macroautophagy marker in ovarian CSCs. In A2780 and SKOV3 ovarian CSCs, LAMP2A and heat shock protein 70 (HSC70) exhibited higher expression levels than in normal adherent cells. Our results showed that the silencing of the LAMP2A gene resulted in reduced sphere formation and enhanced GLUT5 expression in ovarian CSCs. Moreover, the treatment with fructose reduced sphere formation and enhanced the expression levels of LAMP2A, SOX2, and OCT4 in ovarian CSCs. The KEGG functional analysis revealed that differentially expressed genes were enriched in the ferroptosis pathway in A2780-spheroid (SP) cells after treatment with fructose. In A2780-SP and SKOV3-SP cells, the level of SLC7A11 decreased whereas FTH increased after treatment with fructose. Taken together, our results suggest that CMA is mediated in CSCs via fructose metabolism.
Collapse
Affiliation(s)
- Eun Jung Sohn
- College of Medicine, Pusan National University, Yangsan, Republic of Korea.; Inje University, 197 Injero, Gimhae 50834, Republic of Korea.
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sec-Ok Oh
- Department of Anatomy, School of Medicine, Yangsan, Republic of Korea; Korea School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jin-Young Kim
- The School of Korean Medicine Pusan National University, Yangsan 50612, Republic of Korea; Korea Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
17
|
Nieddu V, Melocchi V, Battistini C, Franciosa G, Lupia M, Stellato C, Bertalot G, Olsen JV, Colombo N, Bianchi F, Cavallaro U. Matrix Gla Protein drives stemness and tumor initiation in ovarian cancer. Cell Death Dis 2023; 14:220. [PMID: 36977707 PMCID: PMC10050398 DOI: 10.1038/s41419-023-05760-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
Ovarian cancer (OC) displays the highest mortality among gynecological tumors, mainly due to early peritoneal dissemination, the high frequency of tumor relapse following primary debulking, and the development of chemoresistance. All these events are thought to be initiated and sustained by a subpopulation of neoplastic cells, termed ovarian cancer stem cells (OCSC), that are endowed with self-renewing and tumor-initiating properties. This implies that interfering with OCSC function should offer novel therapeutic perspectives to defeat OC progression. To this aim, a better understanding of the molecular and functional makeup of OCSC in clinically relevant model systems is essential. We have profiled the transcriptome of OCSC vs. their bulk cell counterpart from a panel of patient-derived OC cell cultures. This revealed that Matrix Gla Protein (MGP), classically known as a calcification-preventing factor in cartilage and blood vessels, is markedly enriched in OCSC. Functional assays showed that MGP confers several stemness-associated traits to OC cells, including a transcriptional reprogramming. Patient-derived organotypic cultures pointed to the peritoneal microenvironment as a major inducer of MGP expression in OC cells. Furthermore, MGP was found to be necessary and sufficient for tumor initiation in OC mouse models, by shortening tumor latency and increasing dramatically the frequency of tumor-initiating cells. Mechanistically, MGP-driven OC stemness was mediated by the stimulation of Hedgehog signaling, in particular through the induction of the Hedgehog effector GLI1, thus highlighting a novel MGP/Hedgehog pathway axis in OCSC. Finally, MGP expression was found to correlate with poor prognosis in OC patients, and was increased in tumor tissue after chemotherapy, supporting the clinical relevance of our findings. Thus, MGP is a novel driver in OCSC pathophysiology, with a major role in stemness and in tumor initiation.
Collapse
Affiliation(s)
- V Nieddu
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - V Melocchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - C Battistini
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - G Franciosa
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - M Lupia
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - C Stellato
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - G Bertalot
- Unità Operativa Multizonale di Anatomia Patologica, APSS, Trento, Italy
- Centre for Medical Sciences - CISMed, University of Trento, Trento, Italy
| | - J V Olsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - N Colombo
- Division of Gynecologic Oncology, European Institute of Oncology IRCSS, Milan, Italy
- University of Milan-Bicocca, Milan, Italy
| | - F Bianchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - U Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy.
| |
Collapse
|
18
|
Han S, Chen X, Li Z. Innate Immune Program in Formation of Tumor-Initiating Cells from Cells-of-Origin of Breast, Prostate, and Ovarian Cancers. Cancers (Basel) 2023; 15:757. [PMID: 36765715 PMCID: PMC9913549 DOI: 10.3390/cancers15030757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Tumor-initiating cells (TICs), also known as cancer stem cells (CSCs), are cancer cells that can initiate a tumor, possess self-renewal capacity, and can contribute to tumor heterogeneity. TICs/CSCs are developed from their cells-of-origin. In breast, prostate, and ovarian cancers, progenitor cells for mammary alveolar cells, prostate luminal (secretory) cells, and fallopian tube secretory cells are the preferred cellular origins for their corresponding cancer types. These luminal progenitors (LPs) express common innate immune program (e.g., Toll-like receptor (TLR) signaling)-related genes. Microbes such as bacteria are now found in breast, prostate, and fallopian tube tissues and their corresponding cancer types, raising the possibility that their LPs may sense the presence of microbes and trigger their innate immune/TLR pathways, leading to an inflammatory microenvironment. Crosstalk between immune cells (e.g., macrophages) and affected epithelial cells (e.g., LPs) may eventually contribute to formation of TICs/CSCs from their corresponding LPs, in part via STAT3 and/or NFκB pathways. As such, TICs/CSCs can inherit expression of innate-immunity/TLR-pathway-related genes from their cells-of-origin; the innate immune program may also represent their unique vulnerability, which can be explored therapeutically (e.g., by enhancing immunotherapy via augmenting TLR signaling).
Collapse
Affiliation(s)
- Sen Han
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Xueqing Chen
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Izycka N, Rucinski M, Andrzejewska M, Szubert S, Nowak-Markwitz E, Sterzynska K. The Prognostic Value of Cancer Stem Cell Markers (CSCs) Expression-ALDH1A1, CD133, CD44-For Survival and Long-Term Follow-Up of Ovarian Cancer Patients. Int J Mol Sci 2023; 24:ijms24032400. [PMID: 36768723 PMCID: PMC9916537 DOI: 10.3390/ijms24032400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Recurrent disease and treatment-associated chemoresistance are the two main factors accounting for poor clinical outcomes of ovarian cancer (OC) patients. Both can be associated with cancer stem cells (CSCs), which contribute to cancer formation, progression, chemoresistance, and recurrence. Hence, this study investigated whether the expression of known CSC-associated markers ALDH1A, CD44, and CD133 may predict OC patient prognosis. We analyzed their expression in primary epithelial ovarian cancer (EOC) patients using immunohistochemistry and related them to clinicopathological data, including overall survival (OS) and progression-free survival (PFS). Expression of ALDH1A1 was detected in 32%, CD133 in 28%, and CD44 in 33% of cases. While Kaplan-Meier analysis revealed no association of the expression of CD133 and CD44 with PFS and OS, ALDH1A1-positive patients were characterized with both significantly shorter OS (p = 0.00022) and PFS (p = 0.027). Multivariate analysis demonstrated that the expression of ALDH1A1, FIGO stage III-IV, and residual disease after suboptimal debulking or neoadjuvant chemotherapy correlated with shorter OS. The results of this study identify ALDH1A1 as a potential independent prognostic factor of shorter OS and PFS in EOC patients. Therefore, targeting ALDH1A1-positive cancer cells may be a promising therapeutic strategy to influence the disease course and treatment response.
Collapse
Affiliation(s)
- Natalia Izycka
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Malgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Sebastian Szubert
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Karolina Sterzynska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
- Correspondence: ; Tel.: +48-61-8546455
| |
Collapse
|
20
|
Alwosaibai K, Aalmri S, Mashhour M, Ghandorah S, Alshangiti A, Azam F, Selwi W, Gharaibeh L, Alatawi Y, Alruwaii Z, Alsaab HO. PD-L1 is highly expressed in ovarian cancer and associated with cancer stem cells populations expressing CD44 and other stem cell markers. BMC Cancer 2023; 23:13. [PMID: 36604635 PMCID: PMC9814309 DOI: 10.1186/s12885-022-10404-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors, including PD-L1 (programmed death ligand-1) inhibitors have well documented anticancer therapeutic effect in most types of cancers but its use in the treatment of ovarian cancer is not yet proven. The aim of our study is to explore the predictive biomarkers in ovarian cancer and its association with the outcomes. We have investigated the role of PD-L1 expressions in the tumor microenvironment cells including immune cells and cancer stem cells in different types of ovarian cancer. METHODS A total of 119 surgical archived ovarian cancer samples were collected from the pathology department at King Fahad Specialist Hospital, Dammam, Saudi Arabia that included serous carcinomas, clear cell carcinomas, mucinous carcinomas, endometrioid carcinomas, and granulosa cell tumors. Immunohistochemistry (IHC) staining was performed using (i) PD-L1 antibodies to detect PD-L1 expressions; (ii) CD8 and CD4 to detect Tumor Infiltrating Lymphocytes (TILs); and (iii) CD44, LGR5, and ALDH2 to detect stem cell markers. The clinicopathological data were collected from patients' medical record to investigate the association with PD-L1, TILs, and stem cells expressions. RESULTS We report high PD-L1 expressions in 47.8% of ovarian cancer samples. PD-L1 expressions were detected in different types of epithelial ovarian cancer and were not associated with poor prognosis of ovarian cancer. However, determining the expression levels of TILs in the ovarian cancer tissues found that 81% (n = 97) of ovarian cancer samples have TILs that express both of CD8 and CD4 and significantly associated with high PD-L1 expressions. Interestingly, we have found that ovarian cancer tissues with high expressions of PD-L1 were associated with high expressions of stem cells expressing CD44 and LGR5. CONCLUSIONS PD-L1 is highly expressed in the serous type of ovarian carcinomas and the overall expression of PD-L1 is not associated with poor survival rate. Furthermore, PD-L1 expressions are strongly associated with TILs and stem cell markers in ovarian cancer. Inhibiting the PD-L1 using immune checkpoint inhibitors might downregulate stem cell population that known to be associated with cancer recurrence.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- grid.415280.a0000 0004 0402 3867Research Center, Biomedical Research Department, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Salmah Aalmri
- grid.415280.a0000 0004 0402 3867Research Center, Biomedical Research Department, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Miral Mashhour
- grid.415280.a0000 0004 0402 3867Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Salim Ghandorah
- grid.415280.a0000 0004 0402 3867Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Abdulraheem Alshangiti
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Faisal Azam
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Waleed Selwi
- grid.415280.a0000 0004 0402 3867Department of Medical Oncology, King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Lubna Gharaibeh
- grid.116345.40000000406441915Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Yasser Alatawi
- grid.440760.10000 0004 0419 5685Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Zainab Alruwaii
- Department of Anatomic Pathology, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Hashem O. Alsaab
- grid.412895.30000 0004 0419 5255Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O BOX 11099, Taif, Saudi Arabia
| |
Collapse
|
21
|
Patient-Derived In Vitro Models of Ovarian Cancer: Powerful Tools to Explore the Biology of the Disease and Develop Personalized Treatments. Cancers (Basel) 2023; 15:cancers15020368. [PMID: 36672318 PMCID: PMC9856518 DOI: 10.3390/cancers15020368] [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: 12/02/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Epithelial ovarian cancer (OC) is the most lethal gynecological malignancy worldwide due to a late diagnosis caused by the lack of specific symptoms and rapid dissemination into the peritoneal cavity. The standard of care for OC treatment is surgical cytoreduction followed by platinum-based chemotherapy. While a response to this frontline treatment is common, most patients undergo relapse within 2 years and frequently develop a chemoresistant disease that has become unresponsive to standard treatments. Moreover, also due to the lack of actionable mutations, very few alternative therapeutic strategies have been designed as yet for the treatment of recurrent OC. This dismal clinical perspective raises the need for pre-clinical models that faithfully recapitulate the original disease and therefore offer suitable tools to design novel therapeutic approaches. In this regard, patient-derived models are endowed with high translational relevance, as they can better capture specific aspects of OC such as (i) the high inter- and intra-tumor heterogeneity, (ii) the role of cancer stem cells (a small subset of tumor cells endowed with tumor-initiating ability, which can sustain tumor spreading, recurrence and chemoresistance), and (iii) the involvement of the tumor microenvironment, which interacts with tumor cells and modulates their behavior. This review describes the different in vitro patient-derived models that have been developed in recent years in the field of OC research, focusing on their ability to recapitulate specific features of this disease. We also discuss the possibilities of leveraging such models as personalized platforms to design new therapeutic approaches and guide clinical decisions.
Collapse
|
22
|
Sotnikova TN, Polushkina TV, Danilova NV. [Relationship between PD-L1 expression and tumor stem cell marker CD44 as a promising basis for the development of new approaches to cancer targeted therapy]. Arkh Patol 2023; 85:70-75. [PMID: 38010641 DOI: 10.17116/patol20238506170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Immunotherapy of malignant tumors is a rapidly developing area of oncology. PD-1 is a receptor expressed by activated T-lymphocytes. As a result of its interaction with the ligand (PD-L1 or PD-L2), the activity of T-lymphocytes is inhibited and their apoptosis occurs. Drugs that inhibit the interaction of PD-1 with ligands have an immunostimulatory effect and are effective in the treatment of many types of neoplasms: melanoma, lung cancer, bladder cancer, stomach cancer, various lymphomas, etc. However, response to this treatment is observed only in a narrow cohort of patients. To increase the effectiveness of immunotherapy, combined preparations and nanoparticles are being developed and created to enhance the effect of PD-L1 inhibitors, and containing hyaluronic acid as a ligand for the CD44 protein, which is expressed in many human tumors. However, the issue of co-expression of CD44 and PD-L1 remains poorly understood. This review is devoted to describing the features of co-expression and the mechanisms of interaction between CD44 and PD-L1. Promising directions for the development of new approaches to the immunotherapy of malignant tumors are presented.
Collapse
Affiliation(s)
- T N Sotnikova
- I.V. Davydovsky City Clinical Hospital, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - T V Polushkina
- I.V. Davydovsky City Clinical Hospital, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - N V Danilova
- Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
23
|
Nanotechnology-Based Nucleic Acid Vaccines for Treatment of Ovarian Cancer. Pharm Res 2023; 40:123-144. [PMID: 36376606 PMCID: PMC9663189 DOI: 10.1007/s11095-022-03434-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Anticancer vaccines represent a promising approach for effective treatment of cancer and along with recent advantages of nucleic acid-based vaccines for other diseases form a prospective and potentially efficacious direction of the research, development and clinical applications. Despite the ongoing several clinical trials of mRNA vaccines for the treatment of various types of cancer, to-date no cancer vaccines were approved by the US Food and Drug Administration. The present review analyzes and summarizes major approaches for treating of different forms of ovarian cancer including mRNA-based vaccines as well as nanotechnology-based approaches for their delivery.
Collapse
|
24
|
Endometriosis Stem Cells as a Possible Main Target for Carcinogenesis of Endometriosis-Associated Ovarian Cancer (EAOC). Cancers (Basel) 2022; 15:cancers15010111. [PMID: 36612107 PMCID: PMC9817684 DOI: 10.3390/cancers15010111] [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: 11/27/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Endometriosis is a serious recurrent disease impairing the quality of life and fertility, and being a risk for some histologic types of ovarian cancer defined as endometriosis-associated ovarian cancers (EAOC). The presence of stem cells in the endometriotic foci could account for the proliferative, migrative and angiogenic activity of the lesions. Their phenotype and sources have been described. The similarly disturbed expression of several genes, miRNAs, galectins and chaperones has been observed both in endometriotic lesions and in ovarian or endometrial cancer. The importance of stem cells for nascence and sustain of malignant tumors is commonly appreciated. Although the proposed mechanisms promoting carcinogenesis leading from endometriosis into the EAOC are not completely known, they have been discussed in several articles. However, the role of endometriosis stem cells (ESCs) has not been discussed in this context. Here, we postulate that ESCs may be a main target for the carcinogenesis of EAOC and present the possible sequence of events resulting finally in the development of EAOC.
Collapse
|
25
|
Sher G, Masoodi T, Patil K, Akhtar S, Kuttikrishnan S, Ahmad A, Uddin S. Dysregulated FOXM1 signaling in the regulation of cancer stem cells. Semin Cancer Biol 2022; 86:107-121. [PMID: 35931301 DOI: 10.1016/j.semcancer.2022.07.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/20/2022] [Accepted: 07/31/2022] [Indexed: 01/27/2023]
Abstract
Since the introduction of the cancer stem cell (CSC) paradigm, significant advances have been made in understanding the functional and biological plasticity of these elusive components in malignancies. Endowed with self-renewing abilities and multilineage differentiation potential, CSCs have emerged as cellular drivers of virtually all facets of tumor biology, including metastasis, tumor recurrence/relapse, and drug resistance. The functional and biological characteristics of CSCs, such as self-renewal, cell fate decisions, survival, proliferation, and differentiation are regulated by an array of extracellular factors, signaling pathways, and pluripotent transcriptional factors. Besides the well-characterized regulatory role of transcription factors OCT4, SOX2, NANOG, KLF4, and MYC in CSCs, evidence for the central role of Forkhead box transcription factor FOXM1 in the establishment, maintenance, and functions of CSCs is accumulating. Conventionally identified as a master regulator of the cell cycle, a comprehensive understanding of this molecule has revealed its multifarious oncogenic potential and uncovered its role in angiogenesis, invasion, migration, self-renewal, and drug resistance. This review compiles the large body of literature that has accumulated in recent years that provides evidence for the mechanisms by which FOXM1 expression promotes stemness in glioblastoma, breast, colon, ovarian, lung, hepatic, and pancreatic carcinomas. We have also compiled the data showing the association of stem cell mediators with FOXM1 using TCGA mRNA expression data. Further, the prognostic importance of FOXM1 and other stem cell markers is presented. The delineation of FOXM1-mediated regulation of CSCs can aid in the development of molecularly targeted pharmacological approaches directed at the selective eradication of CSCs in several human malignancies.
Collapse
Affiliation(s)
- Gulab Sher
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar
| | - Kalyani Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
26
|
Therachiyil L, Hussein OJ, Uddin S, Korashy HM. Regulation of the aryl hydrocarbon receptor in cancer and cancer stem cells of gynecological malignancies: An update on signaling pathways. Semin Cancer Biol 2022; 86:1186-1202. [PMID: 36252938 DOI: 10.1016/j.semcancer.2022.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 01/27/2023]
Abstract
Gynecological malignancies are a female type of cancers that affects the reproductive system. Cancer metastasis or recurrence mediated by cellular invasiveness occurs at advanced stages of cancer progression. Cancer Stem Cells (CSCs) enrichment in tumors leads to chemoresistance, which results in cancer mortality. Exposure to environmental pollutants such as polycyclic aromatic hydrocarbons is associated with an increased the risk of CSC enrichment in gynecological cancers. One of the important pathways that mediates the metabolism and bioactivation of these environmental chemicals is the transcription factor, aryl hydrocarbon receptor (AhR). The present review explores the molecular mechanisms regulating the crosstalk and interaction of the AhR with cancer-related signaling pathways, such as apoptosis, epithelial-mesenchymal transition, immune checkpoints, and G-protein-coupled receptors in several gynecological malignancies such as ovarian, uterine, endometrial, and cervical cancers. The review also discusses the potential of targeting the AhR pathway as a novel chemotherapy for gynecological cancers.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Ola J Hussein
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
27
|
Liu X, Zhao Y, Jiao X, Yu Y, Li R, Zeng S, Chi J, Ma G, Huo Y, Li M, Peng Z, Liu J, Zhou Q, Zou D, Wang L, Li Q, Wang J, Yao S, Chen Y, Ma D, Hu T, Gao Q. Does the primary treatment sequence affect post-relapse survival in recurrent epithelial ovarian cancer? A real-world multicentre retrospective study. BJOG 2022; 129 Suppl 2:70-78. [PMID: 36485065 DOI: 10.1111/1471-0528.17329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To explore the impact of the primary treatment sequence (primary debulking surgery, PDS, versus neoadjuvant chemotherapy and interval debulking surgery, NACT-IDS) on post-relapse survival (PRS) and recurrence characteristics of recurrent epithelial ovarian cancer (REOC). DESIGN Real-world retrospective study. SETTING Tertiary hospitals in China. POPULATION A total of 853 patients with REOC at International Federation of Gynaecology and Obstetrics stages IIIC-IV from September 2007 to June 2020. Overall, 377 and 476 patients received NACT-IDS and PDS, respectively. METHODS Propensity score-based inverse probability of treatment weighting (IPTW) was performed to balance the between-group differences. MAIN OUTCOME MEASURES Clinicopathological factors related to PRS. RESULTS The overall median PRS was 29.3 months (95% CI 27.0-31.5 months). Multivariate analysis before and after IPTW adjustment showed that NACT-IDS and residual R1/R2 disease were independent risk factors for PRS (p < 0.05). Patients with diffuse carcinomatosis and platinum-free interval (PFI) ≤ 12 months had a significantly worse PRS (p < 0.001). Logistic regression analysis revealed that NACT-IDS was an independent risk factor for diffuse carcinomatosis (OR 1.36, 95% CI 1.01-1.82, p = 0.040) and PFI ≤ 12 months (OR 1.59, 95% CI 1.08-2.35, p = 0.019). In IPTW analysis, NACT-IDS was still significantly associated with diffuse carcinomatosis (OR 1.29, 95% CI 1.05-1.58, p = 0.015) and PFI ≤ 12 months (OR 1.90, 95% CI 1.52-2.38, p < 0.001). CONCLUSIONS The primary treatment sequence may affect the PRS of patients with REOC by altering the recurrence pattern and PFI duration.
Collapse
Affiliation(s)
- Xingyu Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingjun Zhao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Jiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Yu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruyuan Li
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoqing Zeng
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Chi
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanchen Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yabing Huo
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zikun Peng
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahao Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Dongling Zou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Wang
- Department of Cancer Biology Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Qingshui Li
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Jing Wang
- Hunan Clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youguo Chen
- Department of Gynecology & Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Hu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
MCM2 in human cancer: functions, mechanisms, and clinical significance. Mol Med 2022; 28:128. [PMID: 36303105 PMCID: PMC9615236 DOI: 10.1186/s10020-022-00555-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022] Open
Abstract
Background Aberrant DNA replication is the main source of genomic instability that leads to tumorigenesis and progression. MCM2, a core subunit of eukaryotic helicase, plays a vital role in DNA replication. The dysfunction of MCM2 results in the occurrence and progression of multiple cancers through impairing DNA replication and cell proliferation. Conclusions MCM2 is a vital regulator in DNA replication. The overexpression of MCM2 was detected in multiple types of cancers, and the dysfunction of MCM2 was correlated with the progression and poor prognoses of malignant tumors. According to the altered expression of MCM2 and its correlation with clinicopathological features of cancer patients, MCM2 was thought to be a sensitive biomarker for cancer diagnosis, prognosis, and chemotherapy response. The anti-tumor effect induced by MCM2 inhibition implies the potential of MCM2 to be a novel therapeutic target for cancer treatment. Since DNA replication stress, which may stimulate anti-tumor immunity, frequently occurs in MCM2 deficient cells, it also proposes the possibility that MCM2 targeting improves the effect of tumor immunotherapy.
Collapse
|
29
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
30
|
Drug resistance in NSCLC is associated with tumor micro-environment. Reprod Biol 2022; 22:100680. [PMID: 35926330 DOI: 10.1016/j.repbio.2022.100680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Tumor cell resistance to chemotherapy is the most critical factor that influences the prognosis of cancer patients. It is generally believed that drug resistance is caused by genetic alterations in tumor cells; however, the relationship between drug resistance and the tumor microenvironment (TME) has not been adequately studied. Herein, we successfully identified drug resistance and sensitivity clusters using single-cell transcriptome sequencing data from GSE149383 and established a proportional hazards model to find genes that affected prognosis. The results showed that marker genes between resistant and sensitive clusters were significantly associated with the TME; additionally, the model showed good reliability. Furthermore, we used bulk RNA-seq data to analyze the expression of CD24 and CYP1B1, which revealed little difference in the levels of the two genes in normal and tumor tissues but a significant difference in their expression between drug-resistant and -sensitive cells. In conclusion, our study demonstrated a link between drug resistance and the TME, and we found that CD24 and CYP1B1 may be key regulators of drug resistance development in tumor cells via altering the TME.
Collapse
|
31
|
Antitumor Effect of Si-Jun-Zi Decoction on SGC7901 Gastric Cancer Cells by CMTM2 Activation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4675815. [PMID: 35873650 PMCID: PMC9303151 DOI: 10.1155/2022/4675815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022]
Abstract
The Si-Jun-Zi decoction (SJZ), a traditional Chinese medicine (TCM) formula, is used clinically against multiple malignancies, including gastric cancer (GC). In previous study, we have shown that SJZ plays an anticancer role in SGC7901 cell xenograft mice models. However, the underlying mechanisms are unclear. The objective of this study was to evaluate the effect and mechanism of SJZ on the proliferation, migration, invasion, and cancer stem cell-like properties of GC cells. High-throughput mRNA sequencing analysis was performed to investigate the global alterations in gene expression in xenograft tumors, and 56 significantly differentially expressed genes (43 upregulated and 13 downregulated genes) were identified between the SJZ group and the Model group totally. We focused on CMTM2, which was significantly increased after SJZ intervention, as a candidate target gene of SJZ. The results indicated that CMTM2 expression was elevated in SJZ-treated SGC7901 cells and knocking-down CMTM2 expression partially hampered the inhibitory effects of SJZ on the proliferation, migration, and invasion of GC cells. Moreover, SJZ treatment repressed the spheroid and colony-forming capacity in GC cells, accompanied by downregulation of stem cell markers including SOX2, NANOG, and CD44. CMTM2 knockdown antagonized the effects of SJZ on the cancer stem cell-like properties of SGC7901 cells. Thus, SJZ effectively suppressed the proliferation, migration, invasion, and cancer stem cell-like properties of GC cells in vitro by upregulating CMTM2 expression.
Collapse
|
32
|
Sriramkumar S, Sood R, Huntington TD, Ghobashi AH, Vuong TT, Metcalfe TX, Wang W, Nephew KP, O'Hagan HM. Platinum-induced mitochondrial OXPHOS contributes to cancer stem cell enrichment in ovarian cancer. J Transl Med 2022; 20:246. [PMID: 35641987 PMCID: PMC9153190 DOI: 10.1186/s12967-022-03447-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/18/2022] [Indexed: 01/06/2023] Open
Abstract
Background Platinum based agents—cisplatin and carboplatin in combination with taxanes are used for the treatment of ovarian cancer (OC) patients. However, the majority of OC patients develop recurrent, platinum resistant disease that is uniformly fatal. Platinum treatment enriches for chemoresistant aldehyde dehydrogenase (ALDH) + ovarian cancer stem cells (OCSCs), which contribute to tumor recurrence and disease relapse. Acquired platinum resistance also includes metabolic reprograming and switching to oxidative phosphorylation (OXPHOS). Chemosensitive cells rely on glycolysis while chemoresistant cells have the ability to switch between glycolysis and OXPHOS, depending on which pathway drives a selective advantage for growth and chemoresistance. High expression of genes involved in OXPHOS and high production of mitochondrial ROS are characteristics of OCSCs, suggesting that OCSCs favor OXPHOS over glycolysis. Based on connections between OCSCs, chemoresistance and OXPHOS, we hypothesize that platinum treatment induces changes in metabolism that contribute to platinum-induced enrichment of OCSCs. Methods The effect of cisplatin on mitochondrial activity was assessed by JC1 staining and expression of OXPHOS genes by RT-qPCR. Cisplatin-induced changes in Sirtuin 1 (SIRT1) levels and activity were assessed by western blot. Small molecule inhibitors of mitochondrial complex I and SIRT1 were used to determine if their enzymatic activity contributes to the platinum-induced enrichment of OCSCs. The percentage of ALDH + OCSCs in OC cells and tumor tissue from xenograft models across different treatment conditions was analyzed using ALDEFLUOR assay and flow cytometry. Results We demonstrate that platinum treatment increases mitochondrial activity. Combined treatment of platinum agents and OXPHOS inhibitors blocks the platinum-induced enrichment of ALDH + OCSCs in vitro and in vivo. Furthermore, platinum treatment increases SIRT1 levels and subsequent deacetylase activity, which likely contributes to the increase in platinum-induced mitochondrial activity. Conclusions These findings on metabolic pathways altered by platinum-based chemotherapy have uncovered key targets that can be exploited therapeutically to block the platinum-induced enrichment of OCSCs, ultimately improving the survival of OC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03447-y.
Collapse
Affiliation(s)
- Shruthi Sriramkumar
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Riddhi Sood
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA
| | - Thomas D Huntington
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Ahmed H Ghobashi
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA
| | - Truc T Vuong
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Tara X Metcalfe
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Weini Wang
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Kenneth P Nephew
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA.,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology and Physiology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heather M O'Hagan
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA. .,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA. .,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA. .,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
33
|
Y KN, Perumalsamy NK, Warrier S, Perumalsamy LR, Dharmarajan A. Wnt antagonist as therapeutic targets in ovarian cancer. Int J Biochem Cell Biol 2022; 145:106191. [PMID: 35272015 PMCID: PMC7616886 DOI: 10.1016/j.biocel.2022.106191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 12/28/2022]
Abstract
Ovarian cancer is a fatal malignancy in women with a low survival rate that demands new therapeutic paradigms. Cancer cells acquire various exclusive alterations to proliferate, invade, metastasize, and escape cell death, acting independently of growth-inducing or growth-inhibiting signals. The nature of cellular signaling in tumorigenesis is interwoven. Wnt signaling is an evolutionarily conserved signaling cascade that has been shown to regulate ovarian cancer pathogenesis. The molecular mechanism of Wnt signaling underlying the development of ovarian cancer, drug resistance, and relapse is not completely understood. Extracellularly secreted Wnt signaling inhibitors are crucial regulators of ovarian cancer tumorigenesis and malignant properties of cancer stem cells. Wnt inhibitors arbitrated modifications affecting Wnt pathway proteins on the cell membranes, in the cytoplasm, and in the nucleus have been shown to span essential contributions in the initiation, progression, and chemoresistance of ovarian cancer. Although many extrinsic inhibitors developed targeting the downstream components of the Wnt signaling pathway, investigating the molecular mechanisms of endogenous secreted inhibitors might substantiate prognostic or therapeutic biomarkers development. Given the importance of Wnt signaling in ovarian cancer, more systematic studies combined with clinical studies are requisite to probe the precise mechanistic interactions of Wnt antagonists in ovarian cancer. This review outlines the latest progress on the Wnt antagonists and ovarian cancer-specific regulators such as micro-RNAs, small molecules, and drugs regulating these Wnt antagonists in ovarian tumourigenesis.
Collapse
Affiliation(s)
- Krithicaa Narayanaa Y
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology & Research, Sri Ramachandra Institute of Higher Education and Research (DU), Tamil Nadu 600116, India
| | - Naveen Kumar Perumalsamy
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology & Research, Sri Ramachandra Institute of Higher Education and Research (DU), Tamil Nadu 600116, India
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, School of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560065, India
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology & Research, Sri Ramachandra Institute of Higher Education and Research (DU), Tamil Nadu 600116, India.
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology & Research, Sri Ramachandra Institute of Higher Education and Research (DU), Tamil Nadu 600116, India; Stem Cell and Cancer Biology Laboratory, Curtin University, Perth, WA, Australia; School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia; Curtin Health and Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| |
Collapse
|
34
|
Involvement of Cancer Stem Cells in Chemoresistant Relapse of Epithelial Ovarian Cancer Identified by Transcriptome Analysis. JOURNAL OF ONCOLOGY 2022; 2022:6406122. [PMID: 35401749 PMCID: PMC8991408 DOI: 10.1155/2022/6406122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. Despite the initial resection and chemotherapeutic treatment, relapse is common, which leads to poor survival rates in patients. A primary cause of recurrence is the persistence of ovarian cancer stem cells (OCSCs) with high tumorigenicity and chemoresistance. To achieve a better therapeutic response in EOC relapse, the mechanisms underlying acquired chemoresistance associated with relapse-initiating OCSCs need to be studied. Transcriptomes of both chemosensitive primary and chemoresistant relapse EOC samples were obtained from ICGC OV-AU dataset for differential expression analysis. The upregulated genes were further studied using KEGG and GO analysis. Significantly increased expression of eighteen CSC-related genes was found in chemoresistant relapse EOC groups. Upregulation of the expression in four hub genes including WNT3A, SMAD3, KLF4, and PAX6 was verified in chemoresistant relapse samples via immunohistochemistry staining, which confirmed the existence and enrichment of OCSCs in chemoresistant relapse EOC. KEGG and GO enrichment analysis in microarray expression datasets of isolated OCSCs indicated that quiescent state, increased ability of drug efflux, and enhanced response to DNA damage may have caused the chemoresistance in relapse EOC patients. These findings demonstrated a correlation between OCSCs and acquired chemoresistance and illustrated potential underlying mechanisms of OCSC-initiated relapse in EOC patients. Meanwhile, the differentially expressed genes in OCSCs may serve as novel preventive or therapeutic targets against EOC recurrence in the future.
Collapse
|
35
|
Kong Q, Yan X, Cheng M, Jiang X, Xu L, Shen L, Yu H, Sun L. p62 Promotes the Mitochondrial Localization of p53 through Its UBA Domain and Participates in Regulating the Sensitivity of Ovarian Cancer Cells to Cisplatin. Int J Mol Sci 2022; 23:ijms23063290. [PMID: 35328718 PMCID: PMC8949157 DOI: 10.3390/ijms23063290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
Chemotherapeutic drug-induced p53-dependent crosstalk among tumor cells affects the sensitivity of tumor cells to chemotherapeutic drugs, contributing to chemoresistance. Therefore, pharmacological targeting of p53 may contribute to overcoming drug resistance. The localization of p53 is closely related to its function. Thus, we assessed the effect of p62 on the coordination of p53 mitochondrial localization under chemotherapeutic drug treatment in ovarian cancer cells. We found that the combined use of the proteasome inhibitor epoxomicin and cisplatin led to the accumulation of p53 and sequestosome1(p62) in the mitochondria, downregulated mitochondrial DNA (mtDNA) transcription, inhibited mitochondrial functions, and ultimately promoted apoptosis by enhancing cisplatin sensitivity in ovarian cancer cells. Moreover, the ubiquitin-associated (UBA) domain of p62 was involved in regulating the mitochondrial localization of p53. Our findings suggest that the interaction between p62 and p53 may be a mechanism that determines the fate of tumor cells. In conclusion, p62 coordinated the mitochondrial localization of p53 through its UBA domain, inhibited mtDNA transcription, downregulated mitochondrial function, and promoted ovarian cancer cell death. Our study demonstrates the important role of p53 localization in tumor cell survival and apoptosis, and provides new insights into understanding the anti-tumor mechanism of targeting the ubiquitin–proteasome system in tumor cells.
Collapse
Affiliation(s)
- Qinghuan Kong
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
| | - Xiaoyu Yan
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
| | - Meiyu Cheng
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
| | - Xin Jiang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China;
| | - Long Xu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
| | - Luyan Shen
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
| | - Huimei Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0-431-8561-9485 or +86-0-431-8561-9110 (H.Y. & L.S.)
| | - Liankun Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Q.K.); (X.Y.); (M.C.); (L.X.); (L.S.)
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0-431-8561-9485 or +86-0-431-8561-9110 (H.Y. & L.S.)
| |
Collapse
|
36
|
Lupia M, Melocchi V, Bizzaro F, Lo Riso P, Dama E, Baronio M, Ranghiero A, Barberis M, Bernard L, Bertalot G, Giavazzi R, Testa G, Bianchi F, Cavallaro U. Integrated molecular profiling of patient-derived ovarian cancer models identifies clinically relevant signatures and tumor vulnerabilities. Int J Cancer 2022; 151:240-254. [PMID: 35218560 PMCID: PMC9310611 DOI: 10.1002/ijc.33983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022]
Abstract
High‐grade serous ovarian carcinoma (HGSOC) is a highly aggressive and intractable neoplasm, mainly because of its rapid dissemination into the abdominal cavity, a process that is favored by tumor‐associated peritoneal ascites. The precise molecular alterations involved in HGSOC onset and progression remain largely unknown due to the high biological and genetic heterogeneity of this tumor. We established a set of different tumor samples (termed the As11‐set) derived from a single HGSOC patient, consisting of peritoneal ascites, primary tumor cells, ovarian cancer stem cells (OCSC) and serially propagated tumor xenografts. The As11‐set was subjected to an integrated RNA‐seq and DNA‐seq analysis which unveiled molecular alterations that marked the different types of samples. Our profiling strategy yielded a panel of signatures relevant in HGSOC and in OCSC biology. When such signatures were used to interrogate the TCGA dataset from HGSOC patients, they exhibited prognostic and predictive power. The molecular alterations also identified potential vulnerabilities associated with OCSC, which were then tested functionally in stemness‐related assays. As a proof of concept, we defined PI3K signaling as a novel druggable target in OCSC.
Collapse
Affiliation(s)
- Michela Lupia
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, Milan, Italy
| | - Valentina Melocchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesca Bizzaro
- Laboratory of Tumor Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Pietro Lo Riso
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Elisa Dama
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Micol Baronio
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Massimo Barberis
- Pathology Unit, European Institute of Oncology IRCCS, Milan, Italy
| | - Loris Bernard
- Clinical Genomics Lab, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Bertalot
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Tumor Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milan, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
37
|
Wilczyński JR, Wilczyński M, Paradowska E. Cancer Stem Cells in Ovarian Cancer-A Source of Tumor Success and a Challenging Target for Novel Therapies. Int J Mol Sci 2022; 23:ijms23052496. [PMID: 35269636 PMCID: PMC8910575 DOI: 10.3390/ijms23052496] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells. The aim of this review is to show the most contemporary knowledge concerning the biology of ovarian cancer stem cells (OCSCs) and their impact on chemo-resistance and prognosis in ovarian cancer patients, as well as to present the treatment options targeted exclusively on the OCSCs. The review presents data concerning the role of cancer stem cells in general and then concentrates on OCSCs. The surface and intracellular OCSCs markers and their meaning both for cancer biology and clinical prognosis, signaling pathways specifically activated in OCSCs, the genetic and epigenetic regulation of OCSCs function including the recent studies on the non-coding RNA regulation, cooperation between OCSCs and the tumor microenvironment (ovarian cancer niche) including very specific environment such as ascites fluid, the role of shear stress, autophagy and metabolic changes for the function of OCSCs, and finally mechanisms of OCSCs escape from immune surveillance, are described and discussed extensively. The possibilities of anti-OCSCs therapy both in experimental settings and in clinical trials are presented, including the recent II phase clinical trials and immunotherapy. OCSCs are a unique population of cancer cells showing a great plasticity, self-renewal potential and resistance against anti-cancer treatment. They are responsible for the progression and recurrence of the tumor. Several completed and ongoing clinical trials have tested different anti-OCSCs drugs which, however, have shown unsatisfactory efficacy in most cases. We propose a novel approach to ovarian cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
- Correspondence:
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| |
Collapse
|
38
|
Giordano M, Decio A, Battistini C, Baronio M, Bianchi F, Villa A, Bertalot G, Freddi S, Lupia M, Jodice MG, Ubezio P, Colombo N, Giavazzi R, Cavallaro U. L1CAM promotes ovarian cancer stemness and tumor initiation via FGFR1/SRC/STAT3 signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:319. [PMID: 34645505 PMCID: PMC8513260 DOI: 10.1186/s13046-021-02117-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer stem cells (CSC) have been implicated in tumor progression. In ovarian carcinoma (OC), CSC drive tumor formation, dissemination and recurrence, as well as drug resistance, thus contributing to the high death-to-incidence ratio of this disease. However, the molecular basis of such a pathogenic role of ovarian CSC (OCSC) has been elucidated only to a limited extent. In this context, the functional contribution of the L1 cell adhesion molecule (L1CAM) to OC stemness remains elusive. METHODS The expression of L1CAM was investigated in patient-derived OCSC. The genetic manipulation of L1CAM in OC cells provided gain and loss-of-function models that were then employed in cell biological assays as well as in vivo tumorigenesis experiments to assess the role of L1CAM in OC cell stemness and in OCSC-driven tumor initiation. We applied antibody-mediated neutralization to investigate L1CAM druggability. Biochemical approaches were then combined with functional in vitro assays to study the molecular mechanisms underlying the functional role of L1CAM in OCSC. RESULTS We report that L1CAM is upregulated in patient-derived OCSC. Functional studies showed that L1CAM promotes several stemness-related properties in OC cells, including sphere formation, tumor initiation and chemoresistance. These activities were repressed by an L1CAM-neutralizing antibody, pointing to L1CAM as a druggable target. Mechanistically, L1CAM interacted with and activated fibroblast growth factor receptor-1 (FGFR1), which in turn induced the SRC-mediated activation of STAT3. The inhibition of STAT3 prevented L1CAM-dependent OC stemness and tumor initiation. CONCLUSIONS Our study implicate L1CAM in the tumorigenic function of OCSC and point to the L1CAM/FGFR1/SRC/STAT3 signaling pathway as a novel driver of OC stemness. We also provide evidence that targeting this pathway can contribute to OC eradication.
Collapse
Affiliation(s)
- Marco Giordano
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Alessandra Decio
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Chiara Battistini
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Micol Baronio
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Fabrizio Bianchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, FG, Italy
| | - Alessandra Villa
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy.,Philochem AG, Otelfingen, Switzerland
| | - Giovanni Bertalot
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy.,Division of Anatomical Pathology, Santa Chiara Hospital, Trento, Italy
| | - Stefano Freddi
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
| | - Michela Lupia
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Maria Giovanna Jodice
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
| | - Paolo Ubezio
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Nicoletta Colombo
- Division of Gynecologic Oncology, European Institute of Oncology IRCSS, Milan, Italy.,University of Milan-Bicocca, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy.
| |
Collapse
|
39
|
Rodgers RJ, Abbott JA, Walters KA, Ledger WL. Translational Physiology of Anti-Müllerian Hormone: Clinical Applications in Female Fertility Preservation and Cancer Treatment. Front Endocrinol (Lausanne) 2021; 12:689532. [PMID: 34557157 PMCID: PMC8454407 DOI: 10.3389/fendo.2021.689532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background Whilst the ability of AMH to induce the regression of the Müllerian ducts in the male fetus is well appreciated, AMH has additional biological actions in relation to steroid biosynthesis and ovarian follicle dynamics. An understanding of the physiology of AMH illuminates the potential therapeutic utility of AMH to protect the ovarian reserve during chemotherapy and in the treatment of female malignancies. The translation of the biological actions of AMH into clinical applications is an emerging focus of research, with promising preliminary results. Objective and Rationale Studies indicate AMH restrains primordial follicle development, thus administration of AMH during chemotherapy may protect the ovarian reserve by preventing the mass activation of primordial follicles. As AMH induces regression of tissues expressing the AMH receptor (AMHRII), administration of AMH may inhibit growth of malignancies expressing AMHR II. This review evaluates the biological actions of AMH in females and appraises human clinical applications. Search Methods A comprehensive search of the Medline and EMBASE databases seeking articles related to the physiological functions and therapeutic applications of AMH was conducted in July 2021. The search was limited to studies published in English. Outcomes AMH regulates primordial follicle recruitment and moderates sex steroid production through the inhibition of transcription of enzymes in the steroid biosynthetic pathway, primarily aromatase and 17α-hydroxylase/17,20-lyase. Preliminary data indicates that administration of AMH to mice during chemotherapy conveys a degree of protection to the ovarian reserve. Administration of AMH at the time of ovarian tissue grafting has the potential to restrain uncontrolled primordial follicle growth during revascularization. Numerous studies demonstrate AMH induced regression of AMHR II expressing malignancies. As this action occurs via a different mechanism to traditional chemotherapeutic agents, AMH has the capacity to inhibit proliferation of chemo-resistant ovarian cancer cells and cancer stem cells. Wider Implications To date, AMH has not been administered to humans. Data identified in this review suggests administration of AMH would be safe and well tolerated. Administration of AMH during chemotherapy may provide a synchronistic benefit to women with an AMHR II expressing malignancy, protecting the ovarian reserve whilst the cancer is treated by dual mechanisms.
Collapse
Affiliation(s)
- Rachael Jean Rodgers
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
40
|
Nowicki A, Kulus M, Wieczorkiewicz M, Pieńkowski W, Stefańska K, Skupin-Mrugalska P, Bryl R, Mozdziak P, Kempisty B, Piotrowska-Kempisty H. Ovarian Cancer and Cancer Stem Cells-Cellular and Molecular Characteristics, Signaling Pathways, and Usefulness as a Diagnostic Tool in Medicine and Oncology. Cancers (Basel) 2021; 13:cancers13164178. [PMID: 34439332 PMCID: PMC8394875 DOI: 10.3390/cancers13164178] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/04/2021] [Accepted: 08/13/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Ovarian cancer is still a high-risk, metastatic disease, often diagnosed at a late stage. Difficulties in its treatment are associated with high resistance to chemotherapy and recurrence. Responsible for the malignant features of cancer are considered to be cancer stem cells (CSCs), which generate new cells by modifying various signaling pathways. Signaling pathways are crucial for the regulation of epithelial-mesenchymal transition, metastasis, and self-renewal of CSCs. New therapies based on the use of inhibitors that block CSC growth and proliferation signals are being investigated. The current histological classification of ovarian tumors, their epidemiology, and the recent knowledge of ovarian CSCs, with particular emphasis on their molecular basis, are important considerations. Abstract Despite the increasing development of medicine, ovarian cancer is still a high-risk, metastatic disease that is often diagnosed at a late stage. In addition, difficulties in its treatment are associated with high resistance to chemotherapy and frequent relapse. Cancer stem cells (CSCs), recently attracting significant scientific interest, are considered to be responsible for the malignant features of tumors. CSCs, as the driving force behind tumor development, generate new cells by modifying different signaling pathways. Moreover, investigations on different types of tumors have shown that signaling pathways are key to epithelial-mesenchymal transition (EMT) regulation, metastasis, and self-renewal of CSCs. Based on these established issues, new therapies are being investigated based on the use of inhibitors to block CSC growth and proliferation signals. Many reports indicate that CSC markers play a key role in cancer metastasis, with hopes placed in their targeting to block this process and eliminate relapses. Current histological classification of ovarian tumors, their epidemiology, and the most recent knowledge of ovarian CSCs, with particular emphasis on their molecular background, are important aspects for consideration. Furthermore, the importance of signaling pathways involved in tumor growth, development, and metastasis, is also presented.
Collapse
Affiliation(s)
- Andrzej Nowicki
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland;
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (B.K.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Wojciech Pieńkowski
- Division of Perinatology and Women’s Diseases, Poznan University of Medical Sciences, 60-535 Poznan, Poland;
| | - Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Paulina Skupin-Mrugalska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-780 Poznan, Poland;
| | - Rut Bryl
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Paul Mozdziak
- Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (B.K.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland;
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
- Correspondence:
| |
Collapse
|
41
|
Qian J, LeSavage BL, Hubka KM, Ma C, Natarajan S, Eggold JT, Xiao Y, Fuh KC, Krishnan V, Enejder A, Heilshorn SC, Dorigo O, Rankin EB. Cancer-associated mesothelial cells promote ovarian cancer chemoresistance through paracrine osteopontin signaling. J Clin Invest 2021; 131:e146186. [PMID: 34396988 DOI: 10.1172/jci146186] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/25/2021] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer is the leading cause of gynecological malignancy-related deaths, due to its widespread intraperitoneal metastases and acquired chemoresistance. Mesothelial cells are an important cellular component of the ovarian cancer microenvironment that promote metastasis. However, their role in chemoresistance is unclear. Here, we investigated whether cancer-associated mesothelial cells promote ovarian cancer chemoresistance and stemness in vitro and in vivo. We found that osteopontin is a key secreted factor that drives mesothelial-mediated ovarian cancer chemoresistance and stemness. Osteopontin is a secreted glycoprotein that is clinically associated with poor prognosis and chemoresistance in ovarian cancer. Mechanistically, ovarian cancer cells induced osteopontin expression and secretion by mesothelial cells through TGF-β signaling. Osteopontin facilitated ovarian cancer cell chemoresistance via the activation of the CD44 receptor, PI3K/AKT signaling, and ABC drug efflux transporter activity. Importantly, therapeutic inhibition of osteopontin markedly improved the efficacy of cisplatin in both human and mouse ovarian tumor xenografts. Collectively, our results highlight mesothelial cells as a key driver of ovarian cancer chemoresistance and suggest that therapeutic targeting of osteopontin may be an effective strategy for enhancing platinum sensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Jin Qian
- Department of Radiation Oncology
| | | | - Kelsea M Hubka
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Chenkai Ma
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, New South Wales, Australia
| | | | | | | | - Katherine C Fuh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, Missouri, USA
| | - Venkatesh Krishnan
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Oliver Dorigo
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Erinn B Rankin
- Department of Radiation Oncology.,Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| |
Collapse
|
42
|
Wang K, Liu S, Dou Z, Zhang S, Yang X. Loss of Krüppel-like factor 9 facilitates stemness in ovarian cancer ascites-derived multicellular spheroids via Notch1/slug signaling. Cancer Sci 2021; 112:4220-4233. [PMID: 34363722 PMCID: PMC8486214 DOI: 10.1111/cas.15100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/29/2022] Open
Abstract
The ascites that develops in advanced OC, both at diagnosis and upon recurrence, is a rich source of multicellular spheroids/aggregates (MCSs/MCAs), which are the major seeds of tumor cell dissemination within the abdominal cavity. However, the molecular mechanism by which specific ascites-derived tumor cells survive and metastasize remains largely unknown. In this study, we elucidated cancer stem cell (CSC) properties of ascites-derived MCSs, concomitant with enhanced malignancy, induced EMT, and low KLF9 (Krüppel-like factor 9) expression, compared with PTCs. KLF9 was also downregulated in OC cell line-derived spheroids and the CD117+ CD44+ subpopulation in MCSs. Functional experiments demonstrated that KLF9 negatively modulated stem-like properties in OC cells. Mechanistic studies revealed that KLF9 reduced the transcriptional expression of Notch1 by directly binding to the Notch1 promoter, thereby inhibiting the function of slug in a CSL-dependent manner. Clinically, expression of KLF9 was associated with histological grade and loss of KLF9 predicts poor prognosis in OC.
Collapse
Affiliation(s)
- Kun Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Shujie Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Zhiyuan Dou
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Shuo Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Xingsheng Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, China
| |
Collapse
|
43
|
Kim DK, Kim YN, Kim YE, Lee SY, Shin MJ, Do EK, Choi KU, Kim SC, Kim KH, Suh DS, Song P, Kim JH. TRIB2 Stimulates Cancer Stem-Like Properties through Activating the AKT-GSK3β-β-Catenin Signaling Axis. Mol Cells 2021; 44:481-492. [PMID: 34326276 PMCID: PMC8334352 DOI: 10.14348/molcells.2021.0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/11/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Tribbles homolog 2 (TRIB2) is implicated in tumorigenesis and drug resistance in various types of cancers. However, the role of TRIB2 in the regulation of tumorigenesis and drug resistance of cancer stem cells (CSCs) is still elusive. In the present study, we showed increased expression of TRIB2 in spheroid-forming and aldehyde dehydrogenase-positive CSC populations of A2780 epithelial ovarian cancer cells. Short hairpin RNA-mediated silencing of TRIB2 expression attenuates the spheroid-forming, migratory, tumorigenic, and drug-resistant properties of A2780 cells, whereas overexpression of TRIB2 increases the CSC-like characteristics. TRIB2 overexpression induced GSK3β inactivation by augmenting AKT-dependent phosphorylation of GSK3β at Ser9, followed by increasing β-catenin level via reducing the GSK3β-mediated phosphorylation of β-catenin. Treatment of TRIB2-ovexpressed A2780 cells with the phosphoinositide-3-kinase inhibitor LY294002 abrogated TRIB2-stimulated proliferation, migration, drug resistance of A2780 cells. These results suggest a critical role for TRIB2 in the regulation of CSC-like properties by increasing the stability of β-catenin protein via the AKT-GSK3β-dependent pathways.
Collapse
Affiliation(s)
- Dae Kyoung Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Yu Na Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ye Eun Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Seo Yul Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Min Joo Shin
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Eun Kyoung Do
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Kyung-Un Choi
- Department of Pathology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Seung-Chul Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ki-Hyung Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Dong-Soo Suh
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Parkyong Song
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| |
Collapse
|
44
|
Motohara T, Yoshida GJ, Katabuchi H. The hallmarks of ovarian cancer stem cells and niches: Exploring their harmonious interplay in therapy resistance. Semin Cancer Biol 2021; 77:182-193. [PMID: 33812986 DOI: 10.1016/j.semcancer.2021.03.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022]
Abstract
The concept of a "cancer stem cell" has evolved over the past decades, and research on cancer stem cell biology has entered into a stage of remarkable progress. Cancer stem cells are a major determining factor contributing to the establishment of phenotypic and functional intratumoral heterogeneity in synchronization with their surrounding "cancer stem cell niches." They serve as the driving force for cancer initiation, metastasis, and therapeutic resistance in various types of malignancies. In verity, reciprocal interplay between ovarian cancer stem cells and their niches involves a complex but ingeniously orchestrated tumor microenvironment within the intraperitoneal milieu and especially contribute to chemotherapy resistance in patients with advanced ovarian cancer. Herein, we review the principles of our current understanding of the biological features of ovarian cancer stem cells, focusing mainly on the precise mechanisms underlying acquired chemotherapy resistance. Furthermore, we highlight the specific roles of various cancer-associated stromal and immune cells in creating possible cancer stem cell niches that regulate ovarian cancer stemness.
Collapse
Affiliation(s)
- Takeshi Motohara
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto City, Kumamoto, 860-8556, Japan.
| | - Go J Yoshida
- Department of Immunological Diagnosis, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hidetaka Katabuchi
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto City, Kumamoto, 860-8556, Japan
| |
Collapse
|
45
|
Robinson M, Gilbert SF, Waters JA, Lujano-Olazaba O, Lara J, Alexander LJ, Green SE, Burkeen GA, Patrus O, Sarwar Z, Holmberg R, Wang C, House CD. Characterization of SOX2, OCT4 and NANOG in Ovarian Cancer Tumor-Initiating Cells. Cancers (Basel) 2021; 13:cancers13020262. [PMID: 33445692 PMCID: PMC7828139 DOI: 10.3390/cancers13020262] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
The identification of tumor-initiating cells (TICs) has traditionally relied on surface markers including CD133, CD44, CD117, and the aldehyde dehydrogenase (ALDH) enzyme, which have diverse expression across samples. A more reliable indication of TICs may include the expression of embryonic transcription factors that support long-term self-renewal, multipotency, and quiescence. We hypothesize that SOX2, OCT4, and NANOG will be enriched in ovarian TICs and may indicate TICs with high relapse potential. We evaluated a panel of eight ovarian cancer cell lines grown in standard 2-D culture or in spheroid-enriching 3-D culture, and correlated expression with growth characteristics, TIC marker expression, and chemotherapy resistance. RNA-sequencing showed that cell cycle regulation pathways involving SOX2 were elevated in 3-D conditions. HGSOC lines had longer doubling-times, greater chemoresistance, and significantly increased expression of SOX2, OCT4, and NANOG in 3-D conditions. CD117+ or ALDH+/CD133+ cells had increased SOX2, OCT4, and NANOG expression. Limiting dilution in in vivo experiments implicated SOX2, but not OCT4 or NANOG, with early tumor-initiation. An analysis of patient data suggested a stronger role for SOX2, relative to OCT4 or NANOG, for tumor relapse potential. Overall, our findings suggest that SOX2 may be a more consistent indicator of ovarian TICs that contribute to tumor repopulation following chemotherapy. Future studies evaluating SOX2 in TIC biology will increase our understanding of the mechanisms that drive ovarian cancer relapse.
Collapse
Affiliation(s)
- Mikella Robinson
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Samuel F. Gilbert
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Jennifer A. Waters
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Omar Lujano-Olazaba
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Jacqueline Lara
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Logan J. Alexander
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Samuel E. Green
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Gregory A. Burkeen
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Omid Patrus
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Zinia Sarwar
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Ryne Holmberg
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Christine Wang
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
| | - Carrie D. House
- Biology Department, San Diego State University, San Diego, CA 92106, USA; (M.R.); (S.F.G.); (J.A.W.); (O.L.-O.); (J.L.); (L.J.A.); (S.E.G.); (G.A.B.); (O.P.); (Z.S.); (R.H.); (C.W.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Correspondence: ; Tel.: +1-(619)-594-3053
| |
Collapse
|
46
|
Hidayat YM, Munizar, Harsono AB, Winarno GNA, Hasanuddin, Salima S. Chemokine Ligand 5 to Predict Optimal Cytoreduction in Ovarian Cancer. Int J Gen Med 2020; 13:1201-1206. [PMID: 33244258 PMCID: PMC7685389 DOI: 10.2147/ijgm.s280858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose The ultimate goal of cytoreduction surgery is the complete removal of all visible tumors (complete cytoreductive surgery) or tumor residues <1 cm (optimal cytoreduction surgery). Following cytoreduction surgery in ovarian cancer, tumor residue is one of the most important prognostic factors. Oncologists strive to be able to predict the outcome of cytoreduction surgery during the presurgical period. The purpose of this study was to assess CCL5 as a modality for determining whether a patient could perform optimal cytoreduction surgery or not. Materials and Methods This was an observational, analytic, and cross-sectional study of patients with ovarian cancer who underwent surgery at the Dr. Hasan Sadikin Bandung from 2019 to 2020. All of the patients had stage I–IV disease based on the International Federation of Gynecology and Obstetrics (FIGO) score. Results In total, 72 patients were enrolled in this study, 31 of whom underwent suboptimal cytoreduction surgery and 41 underwent optimal cytoreduction surgery. The mean serum CCL5 level at suboptimal cytoreduction was 70,920.87 ± 36,362.966, while that at optimal cytoreduction was 43,244.95 ± 21,983.887. CCL5, as a predictor of suboptimal cytoreduction surgery, had a sensitivity of 61.3%, a specificity of 68.3%, and an accuracy of 65.7% (p = 0.012). Conclusion Preoperative CCL5 serum levels can predict suboptimal cytoreduction surgery outcomes in patients with ovarian cancer.
Collapse
Affiliation(s)
- Yudi Mulyana Hidayat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Munizar
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Ali Budi Harsono
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Hasanuddin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Syiah Kuala, Aceh, Indonesia
| | - Siti Salima
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
47
|
LncRNAs in Ovarian Cancer Progression, Metastasis, and Main Pathways: ceRNA and Alternative Mechanisms. Int J Mol Sci 2020; 21:ijms21228855. [PMID: 33238475 PMCID: PMC7700431 DOI: 10.3390/ijms21228855] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OvCa) develops asymptomatically until it reaches the advanced stages with metastasis, chemoresistance, and poor prognosis. Our review focuses on the analysis of regulatory long non-coding RNAs (lncRNAs) competing with protein-coding mRNAs for binding to miRNAs according to the model of competitive endogenous RNA (ceRNA) in OvCa. Analysis of publications showed that most lncRNAs acting as ceRNAs participate in OvCa progression: migration, invasion, epithelial-mesenchymal transition (EMT), and metastasis. More than 30 lncRNAs turned out to be predictors of survival and/or response to therapy in patients with OvCa. For a number of oncogenic (CCAT1, HOTAIR, NEAT1, and TUG1 among others) and some suppressive lncRNAs, several lncRNA/miRNA/mRNA axes were identified, which revealed various functions for each of them. Our review also considers examples of alternative mechanisms of actions for lncRNAs besides being ceRNAs, including binding directly to mRNA or protein, and some of them (DANCR, GAS5, MALAT1, and UCA1 among others) act by both mechanisms depending on the target protein. A systematic analysis based on the data from literature and Panther or KEGG (Kyoto Encyclopedia of Genes and Genomes) databases showed that a significant part of lncRNAs affects the key pathways involved in OvCa metastasis, EMT, and chemoresistance.
Collapse
|
48
|
Uddin MH, Kim B, Cho U, Azmi AS, Song YS. Association of ALDH1A1-NEK-2 axis in cisplatin resistance in ovarian cancer cells. Heliyon 2020; 6:e05442. [PMID: 33241139 PMCID: PMC7672295 DOI: 10.1016/j.heliyon.2020.e05442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 10/06/2020] [Accepted: 11/03/2020] [Indexed: 01/05/2023] Open
Abstract
Development of acquired resistance to cisplatin (CDDP) is a major obstacle in the treatment of ovarian cancer patients. According to the cancer stem cell (CSC) hypothesis, the recurrence and chemoresistance are presumed to be linked to cancer stem/progenitor cells. Here, we investigated the CSC-like phenotypes and mechanism of chemoresistance in CDDP resistant ovarian cancer cells. A well-established CDDP sensitive ovarian cancer cell line A2780 and its resistant population A2780-Cp were used. We also developed a supra resistant population (SKOV3-Cp) from a naturally CDDP resistant cell line SKOV3. Both resistant/supra resistant cell lines showed significantly higher self-renewal capability than their parental counterparts. They also showed significant resistance to apoptosis and sub-G1 arrest by CDDP treatment. Stem cell marker ALDH1 positivity rates were higher both in A2780-Cp and SKOV3-Cp cell lines than in their counterparts, quantified by Aldefluor assay kit. Hoechst 33342 dye effluxing side populations were increased up to about five folds in A2780-Cp cells and two folds in SKOV3-Cp cells compared to A2780 and SKOV3 cells, respectively. Among major stemness related genes (POU5F1/OCT4, SOX2, NANOG, NES, BMI1, KLF4 and ALDH1A1), ALDH1A1 and KLF4 were significantly overexpressed in both resistant/supra resistant cells. Silencing ALDH1A1 in A2780 and A2780-Cp cells using siRNA greatly reduced the stem cell population and sensitized cells to CDDP. Moreover, silencing of ALDH1A1 reduced the transcript and protein level of its downstream target NEK-2. We also observed the downregulation of ABC transporters (ABCB1/MDR1, ABCG2 and ABCC1/MRP1) either by ALDH1A1 or NEK-2 silencing and upreguation of ABCB1/MDR1 due to the overexpression of NEK-2. Taken together, the present study suggests that stemness gene ALDH1A1 can be involved in CDDP resistance through the upregulation of NEK-2 in ovarian cancer.
Collapse
Affiliation(s)
- Md Hafiz Uddin
- Gynecological Oncology Laboratory, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Oncology, Karmanos Cancer Institute, Wayen State University, Detroit, Michigan 48201, USA
| | - Boyun Kim
- Gynecological Oncology Laboratory, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Untack Cho
- Gynecological Oncology Laboratory, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayen State University, Detroit, Michigan 48201, USA
| | - Yong Sang Song
- Gynecological Oncology Laboratory, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea.,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul, 03080, Republic of Korea
| |
Collapse
|
49
|
Etzerodt A, Moulin M, Doktor TK, Delfini M, Mossadegh-Keller N, Bajenoff M, Sieweke MH, Moestrup SK, Auphan-Anezin N, Lawrence T. Tissue-resident macrophages in omentum promote metastatic spread of ovarian cancer. J Exp Med 2020; 217:133611. [PMID: 31951251 PMCID: PMC7144521 DOI: 10.1084/jem.20191869] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/23/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence.
Collapse
Affiliation(s)
- Anders Etzerodt
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Morgane Moulin
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Thomas Koed Doktor
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | | - Marc Bajenoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Michael H Sieweke
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Regenerative Therapies, TU Dresden, Dresden, Germany
| | - Søren Kragh Moestrup
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Toby Lawrence
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK.,Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
50
|
Huang X, Tang J. Human La Protein: An RNA-Binding Protein Involved in Ovarian Cancer Development and Multidrug Resistance. Onco Targets Ther 2020; 13:10721-10727. [PMID: 33132701 PMCID: PMC7592153 DOI: 10.2147/ott.s269983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Multidrug resistance is the main cause of chemotherapy failure and death in patients with advanced ovarian cancer. Drug resistance is a problem that must be solved to improve the survival rate of patients with advanced ovarian cancer. The RNA-binding protein La and the La-related protein family are highly expressed in various malignant tumors, including ovarian cancer. This article reviews the mechanisms of La protein in tumorigenesis, development, and drug resistance. High La protein expression in tumor cells promotes tumor proliferation, invasion, and migration; disrupts cell cycle; and inhibits tumor cell apoptosis caused by chemotherapeutic drugs through various pathways, resulting in chemotherapy resistance in ovarian cancer. Further study of the role of La protein in ovarian cancer multidrug resistance may be conducive to the development of human La protein-specific inhibitors that suppress ovarian cancer drug resistance.
Collapse
Affiliation(s)
- Xuan Huang
- Department of Pharmacy, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, People's Republic of China
| | - Jing Tang
- Department of Pharmacy, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, People's Republic of China
| |
Collapse
|