1
|
Malek Mohammadi M, Rismanchi H, Esmailzadeh S, Farahani A, Hedayati N, Alimohammadi M, Mafi A, Farahani N, Hushmandi K. The emerging role of circular RNAs in cisplatin resistance in ovarian cancer: From molecular mechanism to future potential. Noncoding RNA Res 2024; 9:1280-1291. [PMID: 39040815 PMCID: PMC11261309 DOI: 10.1016/j.ncrna.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/05/2024] [Accepted: 05/19/2024] [Indexed: 07/24/2024] Open
Abstract
Ovarian cancer (OC) is the most common cause of death in female cancers. The prognosis of OC is very poor due to delayed diagnosis and identification of most patients in advanced stages, metastasis, recurrence, and resistance to chemotherapy. As chemotherapy with platinum-based drugs such as cisplatin (DDP) is the main treatment in most OC cases, resistance to DDP is an important obstacle to achieving satisfactory therapeutic efficacy. Consequently, knowing the different molecular mechanisms involved in resistance to DDP is necessary to achieve new therapeutic approaches. According to numerous recent studies, non-coding RNAs (ncRNAs) could regulate proliferation, differentiation, apoptosis, and chemoresistance in many cancers, including OC. Most of these ncRNAs are released by tumor cells into human fluid, allowing them to be used as tools for diagnosis. CircRNAs are ncRNA family members that have a role in the initiation, progression, and chemoresistance regulation of various cancers. In the current study, we investigated the roles of several circRNAs and their signaling pathways on OC progression and also on DDP resistance during chemotherapy.
Collapse
Affiliation(s)
| | - Hamidreza Rismanchi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shakiba Esmailzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Farahani
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Pan X, Shi X, Zhang H, Chen Y, Zhou J, Shen F, Wang J, Jiang R. Exosomal miR-4516 derived from ovarian cancer stem cells enhanced cisplatin tolerance in ovarian cancer by inhibiting GAS7. Gene 2024; 927:148738. [PMID: 38955306 DOI: 10.1016/j.gene.2024.148738] [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: 03/22/2024] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer (OC) is a devastating disease for women, with chemotherapy resistance taking the lead. Cisplatin has been the first-line therapy for OC for a long time. However, the resistance of OC to cisplatin is an important impediment to its efficacy. Mounting studies showed that ovarian cancer stem cells (OCSCs) affected chemotherapy resistance by secreting exosomes. MicroRNAs (miRNAs) play important roles in exosomes secreted by OCSCs. Here, through the analysis of GEO database (GSE107155) combined with RT-qPCR of OC-related cells/clinical tissues, it was found that hsa-miR-4516 (miR-4516) was significantly up-regulated in OCSCs. Then, OCSCs-derived exosomes were isolated and identified, and it was observed the influence of exosomes on the chemoresistance in SKOV3/cisplatin (SKOV3/DDP) cells. These results manifested that OCSCs-mediated exosomes facilitated the chemoresistance of SKOV3/DDP cells by delivering miR-4516 into them. Growth arrest-specific 7 (GAS7), a downstream target of miR-4516, was determined by bioinformatics prediction combined with molecular biological detection. Next, we up-regulated GAS7 expression and discovered that the promotion of chemoresistance in SKOV3/DDP cells by OCSCs-derived exosomes was significantly impaired. Finally, the mice tumor model of SKOV3/DDP cells was built to estimate the effect of GAS7 over-expression on OC growth. The results showed that GAS7 inhibited the chemoresistance of OC in vivo. In conclusion, our experiments suggested that OCSCs-derived exosomes enhanced OC cisplatin resistance by suppressing GAS7 through the delivery of miR-4516. This study provides a possible target for the treatment of OC DDP resistance.
Collapse
Affiliation(s)
- Xin Pan
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Xiu Shi
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Hong Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - YouGuo Chen
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - JinHua Zhou
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - FangRong Shen
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Juan Wang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Rong Jiang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China.
| |
Collapse
|
3
|
Begum S, Irvin SD, Cox CK, Huang Z, Wilson JJ, Monroe JD, Gibert Y. Anti-ovarian cancer migration and toxicity characteristics of a platinum(IV) pro-drug with axial HDAC inhibitor ligands in zebrafish models. Invest New Drugs 2024:10.1007/s10637-024-01479-3. [PMID: 39433643 DOI: 10.1007/s10637-024-01479-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
Ovarian cancer is the fifth leading cause of cancer related death in the United States. Cisplatin is a platinum-based anti-cancer drug used against ovarian cancer that enters malignant cells and then damages DNA causing cell death. Typically, ovarian cancer cells become resistant to cisplatin making it necessary to increase subsequent dosage, which usually leads to side-effects including irreversible damage to kidney and auditory system tissue. Ovarian cancer resistance is often associated with upregulation of histone deacetylase (HDAC) enzymes that cause DNA to adopt a closed configuration which reduces the ability of cisplatin to target and damage DNA. Compound B, a platinum(IV) complex with two axial phenylbutyrate (PBA) HDAC inhibitor ligands attached to a cisplatin core, can simultaneously inhibit HDAC activity and damage DNA causing decreased cancer cell viability in cisplatin-sensitive (A2780) and -resistant (A2780cis) ovarian cancer cell lines. However, compound B was not previously evaluated in vivo. As simultaneously inhibiting HDAC-mediated resistance with cisplatin treatment could potentiate the platinum drug's effect, we first confirmed the anti-cancer effect of compound B in the A2780 and A2780cis cell lines using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide spectrophotometric assay. Then, we used zebrafish embryo and transgenic animal models to comparatively analyze the effect of cisplatin, compound B, and controls on general organismal, auditory, and renal system toxicity, and cancer metastasis. We found that lower dosages of compound B (0.3 or 0.6 µM) than of cisplatin (2.0 µM) could cause similar or decreased levels of general, auditory, and renal tissue toxicity, and at 0.6 µM, compound B reduces cancer metastasis more than 2.0 µM cisplatin.
Collapse
Affiliation(s)
- Salma Begum
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Scheldon D Irvin
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Carol K Cox
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106-9510, USA
| | - Jerry D Monroe
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
4
|
Plesselova S, Calar K, Axemaker H, Sahly E, Bhagia A, Faragher JL, Fink DM, de la Puente P. Multicompartmentalized Microvascularized Tumor-on-a-Chip to Study Tumor-Stroma Interactions and Drug Resistance in Ovarian Cancer. Cell Mol Bioeng 2024; 17:345-367. [PMID: 39513004 PMCID: PMC11538101 DOI: 10.1007/s12195-024-00817-y] [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: 02/18/2024] [Accepted: 07/26/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction The majority of ovarian cancer (OC) patients receiving standard of care chemotherapy develop chemoresistance within 5 years. The tumor microenvironment (TME) is a dynamic and influential player in disease progression and therapeutic response. However, there is a lack of models that allow us to elucidate the compartmentalized nature of TME in a controllable, yet physiologically relevant manner and its critical role in modulating drug resistance. Methods We developed a 3D microvascularized multiniche tumor-on-a-chip formed by five chambers (central cancer chamber, flanked by two lateral stromal chambers and two external circulation chambers) to recapitulate OC-TME compartmentalization and study its influence on drug resistance. Stromal chambers included endothelial cells alone or cocultured with normal fibroblasts or cancer-associated fibroblasts (CAF). Results The tumor-on-a-chip recapitulated spatial TME compartmentalization including vessel-like structure, stromal-mediated extracellular matrix (ECM) remodeling, generation of oxygen gradients, and delayed drug diffusion/penetration from the circulation chamber towards the cancer chamber. The cancer chamber mimicked metastasis-like migration and increased drug resistance to carboplatin/paclitaxel treatment in the presence of CAF when compared to normal fibroblasts. CAF-mediated drug resistance was rescued by ECM targeted therapy. Critically, these results demonstrate that cellular crosstalk recreation and spatial organization through compartmentalization are essential to determining the effect of the compartmentalized OC-TME on drug resistance. Conclusions Our results present a functionally characterized microvascularized multiniche tumor-on-a-chip able to recapitulate TME compartmentalization influencing drug resistance. This technology holds the potential to guide the design of more effective and targeted therapeutic strategies to overcome chemoresistance in OC. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00817-y.
Collapse
Affiliation(s)
- Simona Plesselova
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Kristin Calar
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Hailey Axemaker
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
| | - Emma Sahly
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- St. Olaf College, Northfield, MN USA
| | - Amrita Bhagia
- MD PhD Program, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| | - Jessica L. Faragher
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- MD PhD Program, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD USA
| | - Darci M. Fink
- Department of Chemistry, Biochemistry & Physics, South Dakota State University, Brookings, SD USA
| | - Pilar de la Puente
- Present Address: Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD USA
- Department of Obstetrics and Gynecology, University of South Dakota Sanford School of Medicine, Sioux Falls, SD USA
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD USA
- Flow Cytometry Core, Sanford Research, Sioux Falls, SD USA
| |
Collapse
|
5
|
Croft W, Pounds R, Jeevan D, Singh K, Balega J, Sundar S, Williams A, Ganesan R, Kehoe S, Ott S, Zuo J, Yap J, Moss P. The chromatin landscape of high-grade serous ovarian cancer metastasis identifies regulatory drivers in post-chemotherapy residual tumour cells. Commun Biol 2024; 7:1211. [PMID: 39341888 PMCID: PMC11438996 DOI: 10.1038/s42003-024-06909-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Disease recurrence following chemotherapy is a major clinical challenge in ovarian cancer (OC), but little is known regarding how the tumour epigenome regulates transcriptional programs underpinning chemoresistance. We determine the single cell chromatin accessibility landscape of omental OC metastasis from treatment-naïve and neoadjuvant chemotherapy-treated patients and define the chromatin accessibility profiles of epithelial, fibroblast, myeloid and lymphoid cells. Epithelial tumour cells display open chromatin regions enriched with motifs for the oncogenic transcription factors MEIS and PBX. Post chemotherapy microenvironments show profound tumour heterogeneity and selection for cells with accessible chromatin enriched for TP53, TP63, TWIST1 and resistance-pathway-activating transcription factor binding motifs. An OC chemoresistant tumour subpopulation known to be present prior to treatment, and characterised by stress-associated gene expression, is enriched post chemotherapy. Nuclear receptors RORa, NR2F6 and HNF4G are uncovered as candidate transcriptional drivers of these cells whilst closure of binding sites for E2F2 and E2F4 indicate post-treated tumour having low proliferative capacity. Delineation of the gene regulatory landscape of ovarian cancer cells surviving chemotherapy treatment therefore reveals potential core transcriptional regulators of chemoresistance, suggesting novel therapeutic targets for improving clinical outcome.
Collapse
Affiliation(s)
- W Croft
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| | - R Pounds
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - D Jeevan
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - K Singh
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - J Balega
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - S Sundar
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - A Williams
- Histopathology Department, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - R Ganesan
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Histopathology Department, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - S Kehoe
- Department of Gynaecological Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - S Ott
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - J Zuo
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - J Yap
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - P Moss
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK.
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Birmingham, UK.
| |
Collapse
|
6
|
Geoffroy K, Mullins-Dansereau V, Leclerc-Desaulniers K, Viens M, Bourgeois-Daigneault MC. Oncolytic vesicular stomatitis virus alone or in combination with JAK inhibitors is effective against ovarian cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200826. [PMID: 39006945 PMCID: PMC11246050 DOI: 10.1016/j.omton.2024.200826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024]
Abstract
Therapy-resistant ovarian cancers have a poor prognosis and novel effective treatment options are urgently needed. In this study, we evaluated the therapeutic efficacy of the oncolytic vesicular stomatitis virus (VSV) against a panel of patient-derived ovarian cancer cell lines of all epithelial subtypes. Notably, we found that most of the cell lines were sensitive to VSV virotherapy. With the objective of improving treatment efficacy for the oncolytic virus-resistant cell lines, we tested various combinations with ovarian cancer standard of care drugs: olaparib, carboplatin, paclitaxel, doxorubicin, cyclophosphamide, and gemcitabine. While none of these combinations revealed to be beneficial, further experiments demonstrated that the antiviral interferon pathway was functional in VSV-resistant cell lines. Given that interferons signal through Janus kinase (JAK)-STAT to mediate their antiviral function, we tested combinations of oncolytic VSV with clinically relevant JAK inhibitors. Our results show that combining VSV with various JAK inhibitors, including ruxolitinib, enhances VSV virotherapy and treatment efficacy. Altogether, we show that VSV, either as a stand-alone treatment or in combination with JAK inhibitors provides an effective therapeutic option for ovarian cancer patients.
Collapse
Affiliation(s)
- Karen Geoffroy
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Victor Mullins-Dansereau
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Kim Leclerc-Desaulniers
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Mélissa Viens
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
7
|
Villagomez FR, Lang J, Nunez-Avellaneda D, Behbakht K, Dimmick HL, Webb P, Nephew KP, Neville M, Woodruff ER, Bitler BG. Claudin-4 remodeling of nucleus-cell cycle crosstalk maintains ovarian tumor genome stability and drives resistance to genomic instability-inducing agents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611120. [PMID: 39282307 PMCID: PMC11398366 DOI: 10.1101/2024.09.04.611120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
During cancer development, the interplay between the nucleus and the cell cycle leads to a state of genomic instability, often accompanied by observable morphological aberrations. These aberrations can be controlled by tumor cells to evade cell death, either by preventing or eliminating genomic instability. In epithelial ovarian cancer (EOC), overexpression of the multifunctional protein claudin-4 is a key contributor to therapy resistance through mechanisms associated with genomic instability. However, the molecular mechanisms underlying claudin-4 overexpression in EOC remain poorly understood. Here, we altered claudin-4 expression and employed a unique claudin-4 targeting peptide (CMP) to manipulate the function of claudin-4. We found that claudin-4 facilitates genome maintenance by linking the nuclear envelope and cytoskeleton dynamics with cell cycle progression. Claudin-4 caused nuclei constriction by excluding lamin B1 and promoting perinuclear F-actin accumulation, associated with remodeling nuclear architecture, thus altering nuclear envelope dynamics. Consequently, cell cycle modifications due to claudin-4 overexpression resulted in fewer cells entering the S-phase and reduced genomic instability. Importantly, disrupting biological interactions of claudin-4 using CMP and forskolin altered oxidative stress cellular response and increased the efficacy of PARP inhibitor treatment. Our data indicate that claudin-4 protects tumor genome integrity by remodeling the crosstalk between the nuclei and the cell cycle, leading to resistance to genomic instability formation and the effects of genomic instability-inducing agents.
Collapse
Affiliation(s)
- Fabian R. Villagomez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Julie Lang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Daniel Nunez-Avellaneda
- Deputy Directorate of Technological Development, Linkage, and Innovation, National Council of Humanities, Sciences, and Technologies, Mexico City, Mexico
| | - Kian Behbakht
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hannah L. Dimmick
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Patricia Webb
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kenneth P. Nephew
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana
- Department of Anatomy, Cell Biology & Physiology, Indiana University, Indianapolis, Indiana
| | - Margaret Neville
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Elizabeth R. Woodruff
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Benjamin G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
8
|
Li X, Guo Y, Xing Z, Gong T, Yang L, Yang T, Chang B, Wang X, Yu B, Guo R. ABT‑737 increases cisplatin sensitivity through the ROS‑ASK1‑JNK MAPK signaling axis in human ovarian cancer cisplatin‑resistant A2780/DDP cells. Oncol Rep 2024; 52:122. [PMID: 39054955 PMCID: PMC11292299 DOI: 10.3892/or.2024.8781] [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: 03/28/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Ovarian cancer is a gynecological malignant tumor with the highest mortality rate, and chemotherapy resistance seriously affects patient therapeutic outcomes. It has been shown that the high expression of anti‑apoptotic proteins Bcl‑2 and Bcl‑xL is closely related to ovarian cancer chemotherapy resistance. Therefore, reducing Bcl‑2 and Bcl‑xL expression levels may be essential for reversing drug resistance in ovarian cancer. ABT‑737 is a BH3‑only protein mimetic, which can effectively inhibit the expression of the anti‑apoptotic proteins Bcl‑xL and Bcl‑2. Although it has been shown that ABT‑737 can increase the sensitivity of ovarian cancer cells to cisplatin, the specific molecular mechanism remains unclear and requires further investigation. In the present study, the results revealed that ABT‑737 can significantly increase the activation levels of JNK and ASK1 induced by cisplatin in A2780/DDP cells, which are cisplatin‑resistant ovarian cancer cells. Inhibition of the JNK and ASK1 pathway could significantly reduce cisplatin cytotoxicity increased by ABT‑737 in A2780/DDP cells, while inhibiting the ASK1 pathway could reduce JNK activation. In addition, it was further determined that ABT‑737 could increase reactive oxygen species (ROS) levels in A2780/DDP cells induced by cisplatin. Furthermore, the inhibition of ROS could significantly reduce JNK and ASK1 activation and ABT‑737‑mediated increased cisplatin cytotoxicity in A2780/DDP cells. Overall, the current data identified that activation of the ROS‑ASK1‑JNK signaling axis plays an essential role in the ability of ABT‑737 to increase cisplatin sensitivity in A2780/DDP cells. Therefore, upregulation the ROS‑ASK1‑JNK signaling axis is a potentially novel molecular mechanism by which ABT‑737 can enhance cisplatin sensitivity of ovarian cancer cells. In addition, the present research can also provide new therapeutic strategies and new therapeutic targets for patients with cisplatin‑resistant ovarian cancer with high Bcl‑2/Bcl‑xL expression patterns.
Collapse
Affiliation(s)
- Xiaoning Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yumeng Guo
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zihan Xing
- Department of Hematology, Linfen Central Hospital, Linfen, Shanxi 041099, P.R. China
| | - Tao Gong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Lijun Yang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Tao Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bingmei Chang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiaoxia Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Rui Guo
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
9
|
Arcieri M, Andreetta C, Tius V, Zapelloni G, Titone F, Restaino S, Vizzielli G. Molecular biology as a driver in therapeutic choices for ovarian cancer. Int J Gynecol Cancer 2024:ijgc-2024-005700. [PMID: 39209430 DOI: 10.1136/ijgc-2024-005700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
The majority of patients with ovarian cancer relapse within 3 years of first line chemotherapy. Therefore, choosing the most appropriate treatment in the recurrence setting has a fundamental role in defining a patient's prognosis. Treatment options include systemic and intra-peritoneal chemotherapy, secondary cytoreductive surgery, and stereotactic body radiotherapy. The best therapeutic choice depends on multiple factors and not only on treatment-free interval. For systemic therapy, prior lines therapy, residual toxicities, comorbidities, performance status, and patient preferences should be taken into account. Secondary cytoreductive surgery can be proposed in patients in which complete tumor resectability can be predicted and in those with oligometastatic disease. Stereotactic body radiotherapy represents a valid alternative to surgery for oligometastatic disease with high local control and minimal toxicity. Current evidence has demonstrated an emerging role of BRCA mutational status and molecular profiling in the impacting response to systemic and local treatments. Therefore, these could provide guidance in the treatment decision process and help identify patients who respond better to poly(ADP-ribose) polymerase (PARP)-inhibitors or immunotherapy or to a combined approach with surgery rather than to platinum-based chemotherapy. Current knowledge in this field could help widen therapeutic options, especially for platinum-resistant patients. In this review, we offer an overview of the state of the art regarding the role of chemotherapy, radiotherapy, and surgery in this setting and their implications in clinical practice and in the treatment decision process, so as to provide the best tailored therapy in patients with recurrent ovarian cancer.
Collapse
Affiliation(s)
- Martina Arcieri
- Clinic of Obstetrics and Gynecology, 'S. Maria della Misericordia' University Hospital, Azienda sanitaria universitaria Friuli Centrale, Udine, Italy
| | - Claudia Andreetta
- Department of Medical Oncology, 'S. Maria della Misericordia' University Hospital, Azienda sanitaria universitaria Friuli Centrale, Udine, Friuli-Venezia Giulia, Italy
| | - Veronica Tius
- Medical Area Department (DAME), in Department of Medicine (DMED), University of Udine, Udine, Friuli-Venezia Giulia, Italy
| | - Giulia Zapelloni
- Medical Area Department (DAME), in Department of Medicine (DMED), University of Udine, Udine, Friuli-Venezia Giulia, Italy
| | - Francesca Titone
- Department of Radiation Oncology, S. Maria della Misericordia' University Hospital, Azienda sanitaria universitaria Friuli Centrale, Udine, Friuli-Venezia Giulia, Italy
| | - Stefano Restaino
- Clinic of Obstetrics and Gynecology, 'S. Maria della Misericordia' University Hospital, Azienda sanitaria universitaria Friuli Centrale, Udine, Italy
- PhD School in Biomedical Sciences, Gender Medicine, Child and Women Health, University of Sassari, Sassari, Sardegna, Italy
| | - Giuseppe Vizzielli
- Clinic of Obstetrics and Gynecology, 'S. Maria della Misericordia' University Hospital, Azienda sanitaria universitaria Friuli Centrale, Udine, Italy
- Medical Area Department (DAME), in Department of Medicine (DMED), University of Udine, Udine, Friuli-Venezia Giulia, Italy
| |
Collapse
|
10
|
Tabrizi L, M. Jones A, Romero-Canelon I, Erxleben A. Multiaction Pt(IV) Complexes: Cytotoxicity in Ovarian Cancer Cell Lines and Mechanistic Studies. Inorg Chem 2024; 63:14958-14968. [PMID: 39083592 PMCID: PMC11323244 DOI: 10.1021/acs.inorgchem.4c01586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
Ovarian cancer has the worst case-to-fatality ratio of all gynecologic malignancies. The main reasons for the high mortality rate are relapse and the development of chemoresistance. In this paper, the cytotoxic activity of two new multiaction platinum(IV) derivatives of cisplatin and oxaliplatin in a panel of ovarian cancer cells is reported. Cis,cis,trans-[Pt(NH3)2Cl2(IPA)(DCA)] (1) and trans-[Pt(DACH)(OX)(IPA)(DCA)] (2) (IPA = indole-3-propionic acid, DCA = dichloroacetate, DACH = 1R,2R-1,2-diaminocyclohexane, OX = oxalate) were synthesized and characterized by elemental analysis, ESI-MS, FT-IR, and 1H, 13C, and195Pt NMR spectroscopy. The biological activity was evaluated in A2780, PEA1, PEA2, SKOV3, SW626, and OVCAR3 cells. Both complexes are potent cytotoxins. Remarkably, complex 2 is 14 times more active in OVCAR3 cells than cisplatin and is able to overcome cisplatin resistance in PEA2 and A2780cis cells, which are models of post-treatment patient-developed and laboratory-induced resistance. This complex also shows activity in 3D cancer models of the A2780 cells. Mechanistic studies revealed that the complexes induce apoptosis via DNA damage and ROS generation.
Collapse
Affiliation(s)
- Leila Tabrizi
- School
of Biological and Chemical Sciences, University
of Galway, Galway H91 TK33, Ireland
- School
of Chemical Sciences, Dublin City University, Dublin D09W6Y4, Ireland
| | - Alan M. Jones
- School
of Pharmacy, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Isolda Romero-Canelon
- School
of Pharmacy, University of Birmingham, Birmingham B15 2TT, U.K.
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Andrea Erxleben
- School
of Biological and Chemical Sciences, University
of Galway, Galway H91 TK33, Ireland
- Synthesis
and Solid State Pharmaceutical Centre (SSPC), Limerick V94 T9PX, Ireland
| |
Collapse
|
11
|
Lacroix A, Farhat R, Robert A, Brenner C, Wiels J, Germini D. The first-in-class pro-apoptotic peptide PEP-010 is effective in monotherapy and in combination with paclitaxel on resistant ovarian adenocarcinoma cell models. Front Pharmacol 2024; 15:1444973. [PMID: 39170706 PMCID: PMC11335512 DOI: 10.3389/fphar.2024.1444973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/11/2024] [Indexed: 08/23/2024] Open
Abstract
Ovarian adenocarcinoma is the gynecological malignancy with the worst prognosis and the highest mortality rate. In the first stages of treatment, chemotherapy results effective, but its prolonged use and high doses lead to the appearance of resistance to treatments and relapse in most patients, representing a major challenge for clinicians. We developed PEP-010, a cell penetrating proapoptotic peptide disrupting the protein-protein interaction between caspase-9 and protein phosphatase 2A, thereby leading to the recovery of their activity in the apoptotic pathway. MTT assay or Annexin-V/Propidium Iodide staining and flow cytometry analysis were used to assess sensitivity to chemotherapies and apoptosis after treatment with PEP-010 in monotherapy or in combination with paclitaxel in ovarian carcinoma cell lines. DNA damage was assessed by immunofluorescence using γH2AX marker. We show here that PEP-010 effectively induces cell death in monotherapy on in up to 55% of cells from ovarian adenocarcinoma cell models resistant to different chemotherapies. Moreover, when used in combination with paclitaxel, one of the therapeutic options for recurrent ovarian carcinoma, PEP-010 showed a beneficial effect leading to the reduction of the IC50 of paclitaxel of 2.2 times and to apoptosis in 87% of cells. The described results suggest the potential therapeutic interest for PEP-010 and lead to the choice of ovarian adenocarcinoma as one of the major indications of the ongoing clinical trial.
Collapse
Affiliation(s)
| | | | - Aude Robert
- UMR1279 INSERM, Université Paris-Saclay, Villejuif, France
| | - Catherine Brenner
- CNRS UMR9018, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris-Saclay, Villejuif, France
| | - Joëlle Wiels
- CNRS UMR9018, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris-Saclay, Villejuif, France
| | | |
Collapse
|
12
|
Shin YB, Choi JY, Yoon MS, Yoo MK, Shin DH, Lee JW. Evaluation of Anticancer Efficacy of D-α-Tocopheryl Polyethylene-Glycol Succinate and Soluplus ® Mixed Micelles Loaded with Olaparib and Rapamycin Against Ovarian Cancer. Int J Nanomedicine 2024; 19:7871-7893. [PMID: 39114180 PMCID: PMC11304412 DOI: 10.2147/ijn.s468935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose Ovarian cancer has the highest mortality rate and lowest survival rate among female reproductive system malignancies. There are treatment options of surgery and chemotherapy, but both are limited. In this study, we developed and evaluated micelles composed of D-α-tocopheryl polyethylene-glycol (PEG) 1000 succinate (TPGS) and Soluplus® (SOL) loaded with olaparib (OLA), a poly(ADP-ribose)polymerase (PARP) inhibitor, and rapamycin (RAPA), a mammalian target of rapamycin (mTOR) inhibitor in ovarian cancer. Methods We prepared micelles containing different molar ratios of OLA and RAPA embedded in different weight ratios of TPGS and SOL (OLA/RAPA-TPGS/SOL) were prepared and physicochemical characterized. Furthermore, we performed in vitro cytotoxicity experiments of OLA, RAPA, and OLA/RAPA-TPGS/SOL. In vivo toxicity and antitumor efficacy assays were also performed to assess the efficacy of the mixed micellar system. Results OLA/RAPA-TPGS/SOL containing a 4:1 TPGS:SOL weight ratio and a 2:3 OLA:RAPA molar ratio showed synergistic effects and were optimized. The drug encapsulation efficiency of this formulation was >65%, and the physicochemical properties were sustained for 180 days. Moreover, the formulation had a high cell uptake rate and significantly inhibited cell migration (**p < 0.01). In the in vivo toxicity test, no toxicity was observed, with the exception of the high dose group. Furthermore, OLA/RAPA-TPGS/SOL markedly inhibited tumor spheroid and tumor growth in vivo. Conclusion Compared to the control, OLA/RAPA-TPGS/SOL showed significant tumor inhibition. These findings lay a foundation for the use of TPGS/SOL mixed micelles loaded with OLA and RAPA in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Ju-Yeon Choi
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Myeong Kyun Yoo
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
- Chungbuk National University Hospital, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| |
Collapse
|
13
|
Giudice E, Salutari V, Sassu CM, Ghizzoni V, Carbone MV, Vertechy L, Fagotti A, Scambia G, Marchetti C. Relacorilant in recurrent ovarian cancer: clinical evidence and future perspectives. Expert Rev Anticancer Ther 2024; 24:649-655. [PMID: 38861580 DOI: 10.1080/14737140.2024.2362178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Relacorilant (CORT125134, Corcept Therapeutics) is a selective glucocorticoid receptor modulator, which reverses the glucocorticoid-mediated anti-apoptotic effects and restores the taxane chemosensitivity in epithelial ovarian cancer cells. Given those preclinical findings, relacorilant is currently under investigation in clinical trials in combination with nab-paclitaxel for the platinum-resistant ovarian cancer setting. AREAS COVERED Already published preclinical and clinical evidence of relacorilant antitumor activity was analyzed and discussed. Ongoing clinical trials registered on clincaltrials.gov were also reported. The review aimed to summarize the status of relacorilant, the mechanism of action, the published and ongoing trials, and its safety and efficacy. EXPERT OPINION Relacorilant combined with nab-paclitaxel, may represent a promising strategy for the treatment of platinum-resistant ovarian cancer patients. After preliminary positive results in terms of clinical efficacy, a randomized phase III trial is ongoing to confirm the findings from the published phase II study.
Collapse
Affiliation(s)
- Elena Giudice
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanda Salutari
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carolina Maria Sassu
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Viola Ghizzoni
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Vittoria Carbone
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Vertechy
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Fagotti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Scambia
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Marchetti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
14
|
Plesselova S, Calar K, Axemaker H, Sahly E, de la Puente P. Multicompartmentalized microvascularized tumor-on-a-chip to study tumor-stroma interactions and drug resistance in ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596456. [PMID: 38853974 PMCID: PMC11160770 DOI: 10.1101/2024.05.29.596456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Introduction The majority of ovarian cancer (OC) patients receiving standard of care chemotherapy develop chemoresistance within 5 years. The tumor microenvironment (TME) is a dynamic and influential player in disease progression and therapeutic response. However, there is a lack of models that allow us to elucidate the compartmentalized nature of TME in a controllable, yet physiologically relevant manner and its critical role in modulating drug resistance. Methods We developed a 3D microvascularized multiniche tumor-on-a-chip formed by five chambers (central cancer chamber, flanked by two lateral stromal chambers and two external circulation chambers) to recapitulate OC-TME compartmentalization and study its influence on drug resistance. Stromal chambers included endothelial cells alone or cocultured with normal fibroblasts or cancer-associated fibroblasts (CAF). Results The tumor-on-a-chip recapitulated spatial TME compartmentalization including vessel-like structure, stromal-mediated extracellular matrix (ECM) remodeling, generation of oxygen gradients, and delayed drug diffusion/penetration from the circulation chamber towards the cancer chamber. The cancer chamber mimicked metastasis-like migration and increased drug resistance to carboplatin/paclitaxel treatment in the presence of CAF when compared to normal fibroblasts. CAF-mediated drug resistance was rescued by ECM targeted therapy. Critically, these results demonstrate that cellular crosstalk recreation and spatial organization through compartmentalization are essential to determining the effect of the compartmentalized OC-TME on drug resistance. Conclusions Our results present a functionally characterized microvascularized multiniche tumor-on-a-chip able to recapitulate TME compartmentalization influencing drug resistance. This technology holds the potential to guide the design of more effective and targeted therapeutic strategies to overcome chemoresistance in OC.
Collapse
|
15
|
Zhang XY, Zhu BC, He M, Dong SS. Proto-oncogene c-Myb potentiates cisplatin resistance of ovarian cancer cells by downregulating lncRNA NKILA and modulating cancer stemness and LIN28A-let7 axis. J Ovarian Res 2024; 17:102. [PMID: 38745302 PMCID: PMC11092198 DOI: 10.1186/s13048-024-01429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
Ovarian cancer is a major gynecological cancer that has poor prognosis associated mainly to its late diagnosis. Cisplatin is an FDA approved ovarian cancer therapy and even though the therapy is initially promising, the patients mostly progress to resistance against cisplatin. The underlying mechanisms are complex and not very clearly understood. Using two different paired cell lines representing cisplatin-sensitive and the cisplatin-resistant ovarian cancer cells, the ES2 and the A2780 parental and cisplatin-resistant cells, we show an elevated proto-oncogene c-Myb in resistant cells. We further show down-regulated lncRNA NKILA in resistant cells with its de-repression in resistant cells when c-Myb is silenced. NKILA negatively correlates with cancer cell and invasion but has no effect on cellular proliferation or cell cycle. C-Myb activates NF-κB signaling which is inhibited by NKILA. The cisplatin resistant cells are also marked by upregulated stem cell markers, particularly LIN28A and OCT4, and downregulated LIN28A-targeted let-7 family miRNAs. Whereas LIN28A and downregulated let-7s individually de-repress c-Myb-mediated cisplatin resistance, the ectopic expression of let-7s attenuates LIN28A effects, thus underlying a c-Myb-NKILA-LIN28A-let-7 axis in cisplatin resistance of ovarian cancer cells that needs to be further explored for therapeutic intervention.
Collapse
Affiliation(s)
- Xue-Yan Zhang
- School of Nursing, Jilin University, Changchun, 130021, Jilin, China
| | - Bo-Chi Zhu
- Department of Neurology, Second Hospital of Jilin University, Changchun, 130022, Jilin, China
| | - Miao He
- Department of Anesthesiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130022, Jilin, China
| | - Shan-Shan Dong
- Department of Anesthesiology, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130022, Jilin, China.
| |
Collapse
|
16
|
Zhang Q, Chen C, Zou X, Wu W, Di Y, Li N, Fu A. Iron promotes ovarian cancer malignancy and advances platinum resistance by enhancing DNA repair via FTH1/FTL/POLQ/RAD51 axis. Cell Death Dis 2024; 15:329. [PMID: 38740757 PMCID: PMC11091064 DOI: 10.1038/s41419-024-06688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
Iron is crucial for cell DNA synthesis and repair, but an excess of free iron can lead to oxidative stress and subsequent cell death. Although several studies suggest that cancer cells display characteristics of 'Iron addiction', an ongoing debate surrounds the question of whether iron can influence the malignant properties of ovarian cancer. In the current study, we initially found iron levels increase during spheroid formation. Furthermore, iron supplementation can promote cancer cell survival, cancer spheroid growth, and migration; vice versa, iron chelators inhibit this process. Notably, iron reduces the sensitivity of ovarian cancer cells to platinum as well. Mechanistically, iron downregulates DNA homologous recombination (HR) inhibitor polymerase theta (POLQ) and relieves its antagonism against the HR repair enzyme RAD51, thereby promoting DNA damage repair to resist chemotherapy-induced damage. Additionally, iron tightly regulated by ferritin (FTH1/FTL) which is indispensable for iron-triggered DNA repair. Finally, we discovered that iron chelators combined with platinum exhibit a synergistic inhibitory effect on ovarian cancer in vitro and in vivo. Our findings affirm the pro-cancer role of iron in ovarian cancer and reveal that iron advances platinum resistance by promoting DNA damage repair through FTH1/FTL/POLQ/RAD51 pathway. Our findings highlight the significance of iron depletion therapy, revealing a promising avenue for advancing ovarian cancer treatment.
Collapse
Affiliation(s)
- Qingyu Zhang
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| | - Caiyun Chen
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Xinxin Zou
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Weifeng Wu
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Yunbo Di
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Ning Li
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, 524023, China.
- Department of Hematology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| | - Aizhen Fu
- Laboratory of Obstetrics and Gynecology, Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
17
|
Nunes M, Bartosch C, Abreu MH, Richardson A, Almeida R, Ricardo S. Deciphering the Molecular Mechanisms behind Drug Resistance in Ovarian Cancer to Unlock Efficient Treatment Options. Cells 2024; 13:786. [PMID: 38727322 PMCID: PMC11083313 DOI: 10.3390/cells13090786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Ovarian cancer is a highly lethal form of gynecological cancer. This disease often goes undetected until advanced stages, resulting in high morbidity and mortality rates. Unfortunately, many patients experience relapse and succumb to the disease due to the emergence of drug resistance that significantly limits the effectiveness of currently available oncological treatments. Here, we discuss the molecular mechanisms responsible for resistance to carboplatin, paclitaxel, polyadenosine diphosphate ribose polymerase inhibitors, and bevacizumab in ovarian cancer. We present a detailed analysis of the most extensively investigated resistance mechanisms, including drug inactivation, drug target alterations, enhanced drug efflux pumps, increased DNA damage repair capacity, and reduced drug absorption/accumulation. The in-depth understanding of the molecular mechanisms associated with drug resistance is crucial to unveil new biomarkers capable of predicting and monitoring the kinetics during disease progression and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Carla Bartosch
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (CI-IPO-Porto), Health Research Network (RISE@CI-IPO-Porto), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Miguel Henriques Abreu
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Alan Richardson
- The School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, Staffordshire, UK;
| | - Raquel Almeida
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Biology Department, Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
18
|
Weidle UH, Birzele F. Deregulated circRNAs in Epithelial Ovarian Cancer With Activity in Preclinical In Vivo Models: Identification of Targets and New Modalities for Therapeutic Intervention. Cancer Genomics Proteomics 2024; 21:213-237. [PMID: 38670587 PMCID: PMC11059596 DOI: 10.21873/cgp.20442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is associated with a dismal prognosis due to development of resistance to chemotherapy and metastasis in the peritoneal cavity and distant organs. In order to identify new targets and treatment modalities we searched the literature for up- and and down-regulated circRNAs with efficacy in preclinical EOC-related in vivo systems. Our search yielded circRNAs falling into the following categories: cisplatin and paclitaxel resistance, transmembrane receptors, secreted factors, transcription factors, RNA splicing and processing factors, RAS pathway-related components, proteolysis and cell-cycle regulation, signaling-related proteins, and circRNAs regulating proteins in additional categories. These findings can be potentially translated by validation and manipulation of the corresponding targets, inhibition of circRNAs with antisense oligonucleotides (ASO), small interfering RNAs (siRNA) or small hairpin RNA (shRNA) or by reconstituting their activity.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
19
|
ALEMZADEH EFFAT, ALLAHQOLI LEILA, MAZIDIMORADI AFROOZ, ALEMZADEH ESMAT, GHASEMI FAHIMEH, SALEHINIYA HAMID, ALKATOUT IBRAHIM. Deciphering resistance mechanisms and novel strategies to overcome drug resistance in ovarian cancer: a comprehensive review. Oncol Res 2024; 32:831-847. [PMID: 38686048 PMCID: PMC11055988 DOI: 10.32604/or.2024.031006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/09/2023] [Indexed: 05/02/2024] Open
Abstract
Ovarian cancer is among the most lethal gynecological cancers, primarily due to the lack of specific symptoms leading to an advanced-stage diagnosis and resistance to chemotherapy. Drug resistance (DR) poses the most significant challenge in treating patients with existing drugs. The Food and Drug Administration (FDA) has recently approved three new therapeutic drugs, including two poly (ADP-ribose) polymerase (PARP) inhibitors (olaparib and niraparib) and one vascular endothelial growth factor (VEGF) inhibitor (bevacizumab) for maintenance therapy. However, resistance to these new drugs has emerged. Therefore, understanding the mechanisms of DR and exploring new approaches to overcome them is crucial for effective management. In this review, we summarize the major molecular mechanisms of DR and discuss novel strategies to combat DR.
Collapse
Affiliation(s)
- EFFAT ALEMZADEH
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - LEILA ALLAHQOLI
- Department of Midwifery, Ministry of Health and Medical Education, Tehran, Iran
| | - AFROOZ MAZIDIMORADI
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - ESMAT ALEMZADEH
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Department of Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - FAHIMEH GHASEMI
- Department of Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - HAMID SALEHINIYA
- Social Determinants of Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - IBRAHIM ALKATOUT
- Kiel School of Gynaecological Endoscopy, Campus Kiel, University Hospitals Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
20
|
Alam S, Giri PK. Novel players in the development of chemoresistance in ovarian cancer: ovarian cancer stem cells, non-coding RNA and nuclear receptors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:6. [PMID: 38434767 PMCID: PMC10905178 DOI: 10.20517/cdr.2023.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/03/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
Ovarian cancer (OC) ranks as the fifth leading factor for female mortality globally, with a substantial burden of new cases and mortality recorded annually. Survival rates vary significantly based on the stage of diagnosis, with advanced stages posing significant challenges to treatment. OC is primarily categorized as epithelial, constituting approximately 90% of cases, and correct staging is essential for tailored treatment. The debulking followed by chemotherapy is the prevailing treatment, involving platinum-based drugs in combination with taxanes. However, the efficacy of chemotherapy is hindered by the development of chemoresistance, both acquired during treatment (acquired chemoresistance) and intrinsic to the patient (intrinsic chemoresistance). The emergence of chemoresistance leads to increased mortality rates, with many advanced patients experiencing disease relapse shortly after initial treatment. This review delves into the multifactorial nature of chemoresistance in OC, addressing mechanisms involving transport systems, apoptosis, DNA repair, and ovarian cancer stem cells (OCSCs). While previous research has identified genes associated with these mechanisms, the regulatory roles of non-coding RNA (ncRNA) and nuclear receptors in modulating gene expression to confer chemoresistance have remained poorly understood and underexplored. This comprehensive review aims to shed light on the genes linked to different chemoresistance mechanisms in OC and their intricate regulation by ncRNA and nuclear receptors. Specifically, we examine how these molecular players influence the chemoresistance mechanism. By exploring the interplay between these factors and gene expression regulation, this review seeks to provide a comprehensive mechanism driving chemoresistance in OC.
Collapse
Affiliation(s)
| | - Pankaj Kumar Giri
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi 110068, India
| |
Collapse
|
21
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
22
|
Tu YP, Hanze E, Zhu F, Lagraauw HM, Sloss CM, Method M, Esteves B, Westin EH, Berkenblit A. Population pharmacokinetics of mirvetuximab soravtansine in patients with folate receptor-α positive ovarian cancer: The antibody-drug conjugate, payload and metabolite. Br J Clin Pharmacol 2024; 90:568-581. [PMID: 37872122 DOI: 10.1111/bcp.15937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
AIMS Mirvetuximab soravtansine is a first-in-class antibody-drug conjugate recently approved for the treatment of folate receptor-α positive ovarian cancer. The aim of this study was to develop a population pharmacokinetic model to describe the concentration-time profiles of mirvetuximab soravtansine, the payload (DM4) and a metabolite (S-methyl-DM4). METHODS Mirvetuximab soravtansine was administered intravenously from 0.15 to 7 mg/kg to 543 patients with predominantly platinum-resistant ovarian cancer in 3 clinical studies, and the plasma drug concentrations were analysed using a nonlinear mixed-effects modelling approach. Stepwise covariate modelling was performed to identify covariates. RESULTS We developed a semi-mechanistic population pharmacokinetic model that included linear and nonlinear routes for the elimination of mirvetuximab soravtansine and a target compartment for the formation and disposition of the payload and metabolite in tumour cells. The clearance and volume of the central compartment were 0.0153 L/h and 2.63 L for mirvetuximab soravtansine, 8.83 L/h and 3.67 L for DM4, and 2.04 L/h and 6.3 L for S-methyl-DM4, respectively. Body weight, serum albumin and age were identified as statistically significant covariates. Exposures in patients with renal or hepatic impairment and who used concomitant cytochrome P450 (CYP) 3A4 inhibitors were estimated. CONCLUSION There is no need for dose adjustment due to covariate effects for mirvetuximab soravtansine administered at the recommended dose of 6 mg/kg based on adjusted ideal body weight. Dose adjustment is not required for patients with mild or moderate renal impairment, mild hepatic impairment, or when concomitant weak and moderate CYP3A4 inhibitors are used.
Collapse
Affiliation(s)
- Ya-Ping Tu
- Clinical Pharmacology, ImmunoGen, Waltham, Massachusetts, USA
| | | | - Fengying Zhu
- Bioanalysis, ImmunoGen, Waltham, Massachusetts, USA
| | | | - Callum M Sloss
- Translational Science, ImmunoGen, Waltham, Massachusetts, USA
| | - Michael Method
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Brooke Esteves
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Eric H Westin
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Anna Berkenblit
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| |
Collapse
|
23
|
Na I, Noh JJ, Kim CK, Lee JW, Park H. Combined radiomics-clinical model to predict platinum-sensitivity in advanced high-grade serous ovarian carcinoma using multimodal MRI. Front Oncol 2024; 14:1341228. [PMID: 38327741 PMCID: PMC10847571 DOI: 10.3389/fonc.2024.1341228] [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: 11/20/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
Introduction We aimed to predict platinum sensitivity using routine baseline multimodal magnetic resonance imaging (MRI) and established clinical data in a radiomics framework. Methods We evaluated 96 patients with ovarian cancer who underwent multimodal MRI and routine laboratory tests between January 2016 and December 2020. The patients underwent diffusion-weighted, contrast-enhanced T1-weighted, and T2-weighted MRI. Subsequently, 293 radiomic features were extracted by manually identifying tumor regions of interest. The features were subjected to the least absolute shrinkage and selection operators, leaving only a few selected features. We built the first prediction model with a tree-based classifier using selected radiomics features. A second prediction model was built by combining the selected radiomic features with four established clinical factors: age, disease stage, initial tumor marker level, and treatment course. Both models were built and tested using a five-fold cross-validation. Results Our radiomics model predicted platinum sensitivity with an AUC of 0.65 using a few radiomics features related to heterogeneity. The second combined model had an AUC of 0.77, confirming the incremental benefits of the radiomics model in addition to models using established clinical factors. Conclusion Our combined radiomics-clinical data model was effective in predicting platinum sensitivity in patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Inye Na
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Joseph J. Noh
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong-Won Lee
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| |
Collapse
|
24
|
Villagomez FR, Lang J, Webb P, Neville M, Woodruff ER, Bitler BG. Claudin-4 modulates autophagy via SLC1A5/LAT1 as a tolerance mechanism for genomic instability in ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576263. [PMID: 38293054 PMCID: PMC10827183 DOI: 10.1101/2024.01.18.576263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Genome instability is key for tumor heterogeneity and derives from defects in cell division and DNA damage repair. Tumors show tolerance for this characteristic, but its accumulation is regulated somehow to avoid catastrophic chromosomal alterations and cell death. Claudin-4 is upregulated and closely associated with genome instability and worse patient outcome in ovarian cancer. This protein is commonly described as a junctional protein participating in processes such as cell proliferation and DNA repair. However, its biological association with genomic instability is still poorly-understood. Here, we used CRISPRi and a claudin mimic peptide (CMP) to modulate the cladudin-4 expression and its function, respectively in in-vitro (high-grade serous carcinoma cells) and in-vivo (patient-derived xenograft in a humanized-mice model) systems. We found that claudin-4 promotes a protective cellular-mechanism that links cell-cell junctions to genome integrity. Disruption of this axis leads to irregular cellular connections and cell cycle that results in chromosomal alterations, a phenomenon associated with a novel functional link between claudin-4 and SLC1A5/LAT1 in regulating autophagy. Consequently, claudin-4's disruption increased autophagy and associated with engulfment of cytoplasm-localized DNA. Furthermore, the claudin-4/SLC1A5/LAT1 biological axis correlates with decrease ovarian cancer patient survival and targeting claudin-4 in-vivo with CMP resulted in increased niraparib (PARPi) efficacy, correlating with increased tumoral infiltration of T CD8+ lymphocytes. Our results show that the upregulation of claudin-4 enables a mechanism that promotes tolerance to genomic instability and immune evasion in ovarian cancer; thus, suggesting the potential of claudin-4 as a translational target for enhancing ovarian cancer treatment.
Collapse
|
25
|
Danziger M, Noble H, Roque DM, Xu F, Rao GG, Santin AD. Microtubule-Targeting Agents: Disruption of the Cellular Cytoskeleton as a Backbone of Ovarian Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1452:1-19. [PMID: 38805122 DOI: 10.1007/978-3-031-58311-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Microtubules are dynamic polymers composed of α- and β-tubulin heterodimers. Microtubules are universally conserved among eukaryotes and participate in nearly every cellular process, including intracellular trafficking, replication, polarity, cytoskeletal shape, and motility. Due to their fundamental role in mitosis, they represent a classic target of anti-cancer therapy. Microtubule-stabilizing agents currently constitute a component of the most effective regimens for ovarian cancer therapy in both primary and recurrent settings. Unfortunately, the development of resistance continues to present a therapeutic challenge. An understanding of the underlying mechanisms of resistance to microtubule-active agents may facilitate the development of novel and improved approaches to this disease.
Collapse
Affiliation(s)
- Michael Danziger
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Helen Noble
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dana M Roque
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fuhua Xu
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gautam G Rao
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
26
|
Xu J, Chen J, Wang D, Li Y, Lian P, Wu X, Yan R. Nafamostat mesylate sensitizes ovarian cancer cells to carboplatin by promoting the ZNF24-mediated inhibition of WNT2B. J Toxicol Sci 2024; 49:467-479. [PMID: 39496384 DOI: 10.2131/jts.49.467] [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: 11/06/2024]
Abstract
Resistance to chemotherapeutic medicines complicates and eventually kills people with ovarian cancer. Nafamostat mesylate (NM) has been used as an adjuvant therapy to enhance chemotherapy sensitivity in several cancers. This study aimed to evaluate the effect of NM on ovarian cancer cells susceptible to carboplatin (CBP) and to determine the underlying mechanism involved. Herein, qRT-PCR, western blot, and IHC were used to analyze mRNA and protein expression. Cell viability and proliferation were measured using the MTT and colony formation assays. Cell migration and invasion were examined using the Transwell assay. Flow cytometry was employed to detect cell apoptosis. The interaction between zinc finger protein 24 (ZNF24) and wingless-type MMTV integration site family member 2b (WNT2B) was validated via the dual-luciferase reporter and Chromatin immunoprecipitation assays. A xenograft nude mouse model was used to assess the effect of NM on CBP sensitivity in vivo. Our results showed that NM intervention inhibited the viability, proliferation, migration, and invasion and facilitated the apoptosis of CBP-resistant ovarian cancer cells. Furthermore, NM sensitized ovarian cancer cells to CBP by upregulating ZNF24. ZNF24 inactivated Wnt/β-catenin signaling by inhibiting the transcription of WNT2B. Additionally, NM enhanced the inhibitory effect of CBP on tumor growth in vivo. Taken together, NM enhanced the CBP sensitivity of ovarian cancer cells by promoting the ZNF24-mediated inactivation of the WNT2B/Wnt/β-catenin axis. These findings suggest a viable treatment approach for improving CBP resistance in ovarian cancer.
Collapse
Affiliation(s)
- Jiehuan Xu
- Changsha Health Vocational College, China
| | - Jianlin Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, China
| | - Dao Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, China
| | - Yaojun Li
- Changsha Health Vocational College, China
| | - Ping Lian
- Changsha Health Vocational College, China
| | - Xiaozhu Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, China
| | - Rong Yan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, China
| |
Collapse
|
27
|
Mairinger E, Wessolly M, Buderath P, Borchert S, Henrich L, Mach P, Steinborn J, Kimming R, Jasani B, Schmid KW, Bankfalvi A, Mairinger FD. Tumor cell cytoplasmic metallothionein expression associates with differential tumor immunogenicity and prognostic outcome in high-grade serous ovarian carcinoma. Front Oncol 2023; 13:1252700. [PMID: 38023247 PMCID: PMC10663300 DOI: 10.3389/fonc.2023.1252700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background The underlying mechanism of high T-cell presence as a favorable prognostic factor in high-grade serous ovarian carcinoma (HGSOC) is not yet understood. In addition to immune cells, various cofactors are essential for immune processes. One of those are metallothioneins (MTs), metal-binding proteins comprising various isoforms. MTs play a role in tumor development and drug resistance. Moreover, MTs influence inflammatory processes by regulating zinc homeostasis. In particular, T-cell function and polarization are particularly susceptible to changes in zinc status. The aim of the present study was to investigate a possible role of MT-mediated immune response and its association with prognostic outcome in ovarian cancer. Methods A retrospective study was conducted on a clinically well-characterized cohort of 24 patients with HGSOC treated at the University Hospital of Essen. Gene expression patterns for anti-cancer immunogenicity-related targets were performed using the NanoString nCounter platform for digital gene expression analysis with the appurtenant PanCancer Immune Profiling panel, consisting of 770 targets and 30 reference genes. Tumor-associated immunohistochemical MT protein expression was evaluated using a semi-quantitative four-tier Immunohistochemistry (IHC) scoring. Results MT immunoexpression was detected in 43% (10/23) of all HGSOC samples. MT immunoexpression levels showed a significant association to survival, leading to prolonged progression-free and overall survival in positively stained tumors. Furthermore, T-cell receptor signaling gene signature showed a strong activation in MT-positive tumors. Activated downstream signaling cascades resulting in elevated interferon-gamma expression with a shift in the balance between T helper cells (TH1 and TH2) could be observed in the MT-positive subgroup. In addition, a higher expression pattern of perforin and several granzymes could be detected, overall suggestive of acute, targeted anti-cancer immune response in MT-positive samples. Conclusion This is the first study combining broad, digital mRNA screening of anti-tumor immune response-associated genes and their relation to MT-I/II in ovarian cancer. MT overexpression is associated with molecular characteristics of an anti-cancer immune response and is a strong prognostic marker in ovarian HGSOC. The observed immune cell activation associated with tumor MT expression comprises but is not limited to T cells and natural killer cells.
Collapse
Affiliation(s)
- Elena Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Michael Wessolly
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Paul Buderath
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Sabrina Borchert
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Larissa Henrich
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Julia Steinborn
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Rainer Kimming
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Bharat Jasani
- Department of Pathology, Targos - A Discovery Life Sciences Company, Kassel, Germany
| | | | - Agnes Bankfalvi
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
28
|
Ayhan S, Hancerliogullari N, Guney G, Gozukucuk M, Caydere M, Guney SS, Tokmak A, Ustun Y. Does the addition of metformin to carboplatin treatment decreases ovarian reserve damage associated with carboplatin usage? J Ovarian Res 2023; 16:184. [PMID: 37660125 PMCID: PMC10474675 DOI: 10.1186/s13048-023-01259-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/13/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND We aimed to determine whether adding metformin to carboplatin treatment would reduce the damage to ovarian reserve associated with carboplatin use. METHODS We included 35 adult female non-pregnant albino Wistar rats approximately three months old, weighing 220-310 g. The rats were divided into five groups of seven rats according to the treatment they received. Carboplatin and salin was given to Group 2, and carboplatin plus metformin was given to Group 3. Group 4 was administered only metformin. Group 5 was administered only salin. Carboplatin was given to Groups 2 and 3 as a single dose on the 15th day, while metformin was given to Groups 3 and 4 during the 28-day experiment. After oophorectomy, histopathologic analyses of primordial, primary, secondary, and tertiary Graff follicles according to the epithelial cells surrounding the oocyte and total follicular number were conducted per section. Serum Anti-Mullerian Hormone (AMH), tissue catalase, and malonyl dialdehyde levels were measured and compared within each group. RESULTS The baseline and 15th-day serum AMH values of the menstrual cycle were compared among the groups, and no statistically significant differences were observed (p > 0.05). Group 3, which was given both carboplatin and metformin, had statistically significantly higher 28th-day AMH levels than Group 2, which was given only carboplatin and saline (p < 0.001). The number of primordial follicles in Group 3 was found to be statistically significantly higher than in Group 2 (p < 0.001). Tissue catalase enzyme levels in Group 3 were statistically significantly higher than in Group 2 (p < 0.001). Tissue malondialdehyde levels in Group 2 were statistically significantly higher than tissue malondialdehyde levels in Groups 3 and 4 (p < 0.001). CONCLUSIONS Metformin may attenuate carboplatin-induced ovarian damage, possibly through its antioxidative effects.
Collapse
Affiliation(s)
- Sevgi Ayhan
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Necati Hancerliogullari
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Gurhan Guney
- Department of Reproductive Endocrinology and Infertility, Balikesir University School of Medicine, Cagis Campus,10145, 10145, Balikesir, Turkey.
| | - Murat Gozukucuk
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Muzaffer Caydere
- Department of Pathology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Sergul Selvi Guney
- Department of Midwifery, Faculty of Health Sciences, Balikesir University, Balikesir, Turkey
| | - Aytekin Tokmak
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Yusuf Ustun
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
29
|
Guan W, Yuan J, Li X, Gao X, Wang F, Liu H, Shi J, Xu G. Cyclin dependent kinase 14 as a paclitaxel-resistant marker regulated by the TGF-β signaling pathway in human ovarian cancer. J Cancer 2023; 14:2538-2551. [PMID: 37670966 PMCID: PMC10475357 DOI: 10.7150/jca.86842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
Cyclin dependent kinase 14 (CDK14) plays a central role in the control of cell proliferation and cell cycle progression. However, the specific function and regulatory mechanism of CDK14 on paclitaxel (PTX) resistance in ovarian cancer (OC) remain unclear. The present study demonstrated that CDK14 was overexpressed in OC tissues and cells at mRNA and protein levels detected by qRT-PCR, Western blot, and immunohistochemistry. Survival analysis showed that elevated CDK14 was related to the poor prognosis of OC patients. Overexpression of CDK14 was correlated with chemoresistance in OC. The expression level of CDK14 was higher in PTX-resistant OC cells (SK3R-PTX and OV3R-PTX) than in their counterpart-sensitive cells (SK-OV-3 and OVCAR-3). Knockdown of CDK14 decreased multidrug resistance 1 (MDR1) and β-catenin expression in SK3R-PTX and OV3R-PTX cells and resensitized OC cells to PTX by decreasing cell proliferation and inducing cell apoptosis. Administration of transforming growth factor (TGF)-β1 decreased CDK14 protein in PTX-resistant OC cells. The inhibitory effect of TGF-β1 on CDK14 expression was abolished in the presence of a TGF-β type I receptor kinase inhibitor (SB-431542). Furthermore, TGF-β signal transducer Smad2 protein directly bound to the region -437 to -446 upstream of the CDK14 transcription start site (TSS), resulting in downregulating the expression of CDK14. These data indicate that CDK14 is a PTX-resistant marker and is regulated by the TGF-β signaling pathway. Targeting CDK14 to enhance the sensitivity of PTX may provide a new therapeutic strategy for reversing the PTX resistance in OC.
Collapse
Affiliation(s)
- Wencai Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jia Yuan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Li
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xuzhu Gao
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Fanchen Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huiqiang Liu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jimin Shi
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
30
|
Adzibolosu N, Alvero AB, Ali-Fehmi R, Gogoi R, Corey L, Tedja R, Chehade H, Gogoi V, Morris R, Anderson M, Vitko J, Lam C, Craig DB, Draghici S, Rutherford T, Mor G. Immunological modifications following chemotherapy are associated with delayed recurrence of ovarian cancer. Front Immunol 2023; 14:1204148. [PMID: 37435088 PMCID: PMC10331425 DOI: 10.3389/fimmu.2023.1204148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Ovarian cancer recurs in most High Grade Serous Ovarian Cancer (HGSOC) patients, including initial responders, after standard of care. To improve patient survival, we need to identify and understand the factors contributing to early or late recurrence and therapeutically target these mechanisms. We hypothesized that in HGSOC, the response to chemotherapy is associated with a specific gene expression signature determined by the tumor microenvironment. In this study, we sought to determine the differences in gene expression and the tumor immune microenvironment between patients who show early recurrence (within 6 months) compared to those who show late recurrence following chemotherapy. Methods Paired tumor samples were obtained before and after Carboplatin and Taxol chemotherapy from 24 patients with HGSOC. Bioinformatic transcriptomic analysis was performed on the tumor samples to determine the gene expression signature associated with differences in recurrence pattern. Gene Ontology and Pathway analysis was performed using AdvaitaBio's iPathwayGuide software. Tumor immune cell fractions were imputed using CIBERSORTx. Results were compared between late recurrence and early recurrence patients, and between paired pre-chemotherapy and post-chemotherapy samples. Results There was no statistically significant difference between early recurrence or late recurrence ovarian tumors pre-chemotherapy. However, chemotherapy induced significant immunological changes in tumors from late recurrence patients but had no impact on tumors from early recurrence patients. The key immunological change induced by chemotherapy in late recurrence patients was the reversal of pro-tumor immune signature. Discussion We report for the first time, the association between immunological modifications in response to chemotherapy and the time of recurrence. Our findings provide novel opportunities to ultimately improve ovarian cancer patient survival.
Collapse
Affiliation(s)
- Nicholas Adzibolosu
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ayesha B. Alvero
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Rouba Ali-Fehmi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Radhika Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Logan Corey
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roslyn Tedja
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Hussein Chehade
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Center of Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Vir Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Robert Morris
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Matthew Anderson
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Julie Vitko
- Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Clarissa Lam
- Department of Gynecologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Douglas B. Craig
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sorin Draghici
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Advaita Corporation, Ann Arbor, MI, United States
- Division of Information and Intelligent Systems, Directorate for Computer and Information Science and Engineering, National Science Foundation, Alexandria, VA, United States
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Gil Mor
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
31
|
Qin M, Zhang C, Li Y. Circular RNAs in gynecologic cancers: mechanisms and implications for chemotherapy resistance. Front Pharmacol 2023; 14:1194719. [PMID: 37361215 PMCID: PMC10285541 DOI: 10.3389/fphar.2023.1194719] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Chemotherapy resistance remains a major challenge in the treatment of gynecologic malignancies. Increasing evidence suggests that circular RNAs (circRNAs) play a significant role in conferring chemoresistance in these cancers. In this review, we summarize the current understanding of the mechanisms by which circRNAs regulate chemotherapy sensitivity and resistance in gynecologic malignancies. We also discuss the potential clinical implications of these findings and highlight areas for future research. CircRNAs are a novel class of RNA molecules that are characterized by their unique circular structure, which confers increased stability and resistance to degradation by exonucleases. Recent studies have shown that circRNAs can act as miRNA sponges, sequestering miRNAs and preventing them from binding to their target mRNAs. This can lead to upregulation of genes involved in drug resistance pathways, ultimately resulting in decreased sensitivity to chemotherapy. We discuss several specific examples of circRNAs that have been implicated in chemoresistance in gynecologic cancers, including cervical cancer, ovarian cancer, and endometrial cancer. We also highlight the potential clinical applications of circRNA-based biomarkers for predicting chemotherapy response and guiding treatment decisions. Overall, this review provides a comprehensive overview of the current state of knowledge regarding the role of circRNAs in chemotherapy resistance in gynecologic malignancies. By elucidating the underlying mechanisms by which circRNAs regulate drug sensitivity, this work has important implications for improving patient outcomes and developing more effective therapeutic strategies for these challenging cancers.
Collapse
|
32
|
Neo SY, Siew YY, Yew HC, He Y, Poh KL, Tsai YC, Ng SL, Tan WX, Chong TI, Lim CSES, Ho SSW, Singh D, Ali A, Linn YC, Tan CH, Seow SV, Koh HL. Effects of Leea indica leaf extracts and its phytoconstituents on natural killer cell-mediated cytotoxicity in human ovarian cancer. BMC Complement Med Ther 2023; 23:79. [PMID: 36899361 PMCID: PMC10007844 DOI: 10.1186/s12906-023-03904-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND The rich biodiversity of medicinal plants and their importance as sources of novel therapeutics and lead compounds warrant further research. Despite advances in debulking surgery and chemotherapy, the risks of recurrence of ovarian cancer and resistance to therapy are significant and the clinical outcomes of ovarian cancer remain poor or even incurable. OBJECTIVE This study aims to investigate the effects of leaf extracts from a medicinal plant Leea indica and its selected phytoconstituents on human ovarian cancer cells and in combination with oxaliplatin and natural killer (NK) cells. METHODS Fresh, healthy leaves of L. indica were harvested and extracted in 70% methanol by maceration. The crude extract was partitioned with n-hexane, dichloromethane and ethyl acetate. Selected extracts and compounds were analyzed for their effects on cell viability of human ovarian cancer cells, NK cell cytotoxicity, and stress ligands expression for NK cell receptors. They were also evaluated for their effects on TNF-α and IL-1β production by enzyme-linked immunosorbent assay in lipopolysaccharide-stimulated human U937 macrophages. RESULTS Leaf extracts of L. indica increased the susceptibility of human ovarian tumor cells to NK cell-mediated cytotoxicity. Treatment of cancer cells with methyl gallate but not gallic acid upregulated the expression of stress ligands. Tumor cells pretreated with combination of methyl gallate and low concentration of oxaliplatin displayed increased levels of stress ligands expression and concomitantly enhanced susceptibility to NK cell-mediated cytolysis. Further, NK cells completely abrogated the growth of methyl gallate-pretreated ovarian cancer cells. The leaf extracts suppressed TNF-α and IL-1β production in human U937 macrophages. Methyl gallate was more potent than gallic acid in down-regulating these cytokine levels. CONCLUSIONS We demonstrated for the first time that leaf extracts of L. indica and its phytoconstituent methyl gallate enhanced the susceptibility of ovarian tumor cells to NK cell cytolysis. These results suggest that the combined effect of methyl gallate, oxaliplatin and NK cells in ovarian cancer cells warrants further investigation, for example for refractory ovarian cancer. Our work is a step towards better scientific understanding of the traditional anticancer use of L. indica.
Collapse
Affiliation(s)
- Soek-Ying Neo
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Yin-Yin Siew
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Hui-Chuing Yew
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Yaqian He
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Keng-Ling Poh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Yi-Chen Tsai
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Shu-Ling Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Wei-Xun Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Teck-Ian Chong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Claire Sophie En-Shen Lim
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Samuel Shan-Wei Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Deepika Singh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| | - Azhar Ali
- Cancer Science Institute of Singapore, 14 Medical Drive, Singapore, 117599 Singapore
| | - Yeh-Ching Linn
- Department of Haematology, Singapore General Hospital, 20 College Road, Singapore, 169856 Singapore
| | - Chay-Hoon Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600 Singapore
| | - See-Voon Seow
- National Cancer Centre Singapore, 11 Hospital Crescent, Singapore, 169610 Singapore
| | - Hwee-Ling Koh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543 Singapore
| |
Collapse
|
33
|
Tossetta G. Metformin Improves Ovarian Cancer Sensitivity to Paclitaxel and Platinum-Based Drugs: A Review of In Vitro Findings. Int J Mol Sci 2022; 23:12893. [PMID: 36361682 PMCID: PMC9654053 DOI: 10.3390/ijms232112893] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
Ovarian cancer is one of the most dangerous gynecologic cancers worldwide, showing a high fatality rate and recurrence due to diagnosis at an advanced stage of the disease and the occurrence of chemoresistance, which weakens the therapeutic effects of the chemotherapeutic treatments. In fact, although paclitaxel and platinum-based drugs (carboplatin or cisplatin) are widely used alone or in combination to treat ovarian cancer, the occurrence of chemoresistance significantly reduces the effects of these drugs. Metformin is a hypoglycemic agent that is commonly used for the treatment of type 2 diabetes mellitus and non-alcoholic fatty liver disease. However, this drug also shows anti-tumor activity, reducing cancer risk and chemoresistance. This review analyzes the current literature regarding the role of metformin in ovarian cancer and investigates what is currently known about its effects in reducing paclitaxel and platinum resistance to restore sensitivity to these drugs.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; ; Tel.: +39-0712206270
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126 Ancona, Italy
| |
Collapse
|
34
|
Wang H, Li S, Yang Y, Zhang L, Zhang Y, Wei T. Perspectives of metal-organic framework nanosystem to overcome tumor drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:954-970. [PMID: 36627891 PMCID: PMC9771744 DOI: 10.20517/cdr.2022.76] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/14/2022] [Accepted: 08/09/2022] [Indexed: 12/23/2022]
Abstract
Cancer is one of the most harmful diseases in the world, which causes huge numbers of deaths every year. Many drugs have been developed to treat tumors. However, drug resistance usually develops after a period of time, which greatly weakens the therapeutic effect. Tumor drug resistance is characterized by blocking the action of anticancer drugs, resisting apoptosis and DNA repair, and evading immune recognition. To tackle tumor drug resistance, many engineered drug delivery systems (DDS) have been developed. Metal-organic frameworks (MOFs) are one kind of emerging and promising nanocarriers for DDS with high surface area and abundant active sites that make the functionalization simpler and more efficient. These features enable MOFs to achieve advantages easily towards other materials. In this review, we highlight the main mechanisms of tumor drug resistance and the characteristics of MOFs. The applications and opportunities of MOF-based DDS to overcome tumor drug resistance are also discussed, shedding light on the future development of MOFs to address tumor drug resistance.
Collapse
Affiliation(s)
- Huafeng Wang
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China.,School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Shi Li
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Yiting Yang
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Lei Zhang
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Yinghao Zhang
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China
| | - Tianxiang Wei
- School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China.,Correspondence to: Dr. Tianxiang Wei, School of Environment, Nanjing Normal University, Nanjing 210023, Jiangsu, China. E-mail:
| |
Collapse
|
35
|
Chen L, Wang J, Liu Q. Long noncoding RNAs as therapeutic targets to overcome chemoresistance in ovarian cancer. Front Cell Dev Biol 2022; 10:999174. [PMID: 36105363 PMCID: PMC9464811 DOI: 10.3389/fcell.2022.999174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/08/2022] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been characterized to play an essential role in ovarian tumorigenesis via controlling a variety of cellular processes, such as cell proliferation, invasion, apoptotic death, metastasis, cell cycle, migration, metabolism, immune evasion, and chemoresistance. The one obstacle for the therapeutic efficacy is due to the development of drug resistance in ovarian cancer patients. Therefore, in this review article, we describe the role of lncRNAs in chemoresistance in ovarian cancer. Moreover, we discuss the molecular mechanism of lncRNAs-involved drug resistance in ovarian cancer. We conclude that lncRNAs could be useful targets to overcome chemoresistance and improve therapeutic outcome in ovarian cancer patients.
Collapse
|
36
|
Restaino S, Wei JJ, Vizzielli G, Martinelli F. Editorial: Molecular influences in therapies in ovarian cancer. Front Oncol 2022; 12:991769. [PMID: 36003794 PMCID: PMC9393694 DOI: 10.3389/fonc.2022.991769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Stefano Restaino
- Department of Maternal and Child Health, Obstetrics and Gynecology Clinic, University-Hospital of Udine, Udine, Italy
- *Correspondence: Stefano Restaino,
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University, School of Medicine, Chicago, IL, United States
| | - Giuseppe Vizzielli
- Department of Medicine, Obstetrics and Gynecology Clinic, University of Udine, Udine, Italy
| | - Fabio Martinelli
- Fondazione Istituto Nazionale Tumori Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| |
Collapse
|