1
|
Zhang F, Zhu T, Gan L, Du Y, Liu L, Ding H, Chen X, Guan Y. FTO triggers NLRP3/GSDMD-dependent pyroptosis to enhance cisplatin-sensitivity in ovarian cancer. Cell Signal 2025; 131:111698. [PMID: 39999914 DOI: 10.1016/j.cellsig.2025.111698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Chemotherapy resistance is still the major impediment to poor prognosis of ovarian cancer (OC). The fat mass and obesity associated protein (FTO), as the first recognized RNA N6-methyladenosine (m6A) demethylase, was dysregulated in various tumors, but its effect on chemoresistance of OC remains unexplored. In this study, FTO expression in tissues and cells was measured by qPCR, IHC, and Western blot. Functional assays were performed, including CCK-8, colony formation, flow cytometry, Transwell assays, and LDH release. Pyroptosis-related proteins were detected by Western blot. MeRIP-qPCR was conducted to measure the m6A level. Finally, the effect of FTO on tumor growth and cisplatin (DDP) sensitivity of OC was checked in a mouse model. We found that FTO was significantly downregulated in OC tissues compared to adjacent normal tissues. In addition, the protein level of FTO was much lower in cisplatin (DDP)-resistant cells than in DDP-sensitive cells. Then, FTO overexpression in DDP-resistant cells decreased IC50 of DDP, increased cell apoptosis, and inhibited cell proliferation, migration, and invasion, whereas FTO silencing cells showed the opposite phenotypes. Moreover, FTO promoted the protein expressions of pyroptosis-related proteins, including NLRP3, cleaved-caspase-1, and GSDMD-NT, to enhance DDP sensitivity, as indicated by lower IC50. Furthermore, FTO activated NLRP3/Caspase-1/GSDMD-mediated pyroptosis in DDP-resistant cells. Mechanically, FTO successfully reduced the total m6A level and NLRP3 mRNA m6A modification to further increase DDP sensitivity in resistant cells. Finally, FTO overexpression promoted DDP-sensitivity and NLRP3 expression to inhibit tumorigenesis in a nude mice xenograft model. Together, our study inferred that FTO triggered NLRP3/Caspase-1/GSDMD-dependent pyroptosis by mediating NLRP3 m6A demethylation to enhance DDP-sensitivity and inhibit the progression of OC. This project indicates that FTO overexpression may be a potential strategy to overcome DDP resistance in OC.
Collapse
Affiliation(s)
- Fubin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Tianhong Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Lei Gan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Yongming Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Lixiao Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Huiqing Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China
| | - Xueqin Chen
- Department of Traditional Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China.
| | - Yutao Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315010, China.
| |
Collapse
|
2
|
L'Espérance K, Madathil S, Ritonja JA, Abrahamowicz M, Ho V, Nicolau B, O'Loughlin J, Koushik A. Trajectories of body fatness in adulthood and the risk of ovarian cancer. Cancer Epidemiol 2025; 96:102814. [PMID: 40245771 DOI: 10.1016/j.canep.2025.102814] [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: 01/27/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND While excess body fatness in older adulthood has been linked to ovarian cancer, the influence of changes in body fatness over time is unclear. This study examined the association between adulthood trajectories of body mass index (BMI), a proxy for body fatness, and ovarian cancer. METHODS In a population-based case-control study (440 cases, 820 controls), we used a group-based trajectory approach to identify BMI trajectories from age 20-70. Using unconditional logistic regression, we estimated adjusted odds ratios (aOR) and 95 % confidence intervals (95 % CI) for the associations between the estimated trajectories and ovarian cancer. RESULTS We identified three distinct BMI trajectories: a normal-stable trajectory, a normal-to-overweight trajectory and an overweight-to-obese trajectory, which included 63.2 %, 31.0 % and 6.8 % of the population, respectively. Multivariable aORs suggested that participants with normal weight at the onset of adulthood who became overweight over their adulthood time did not differ in their risk of ovarian cancer compared to those who maintained a normal weight throughout adulthood (aOR (95 %CI): 0.89 (0.69-1.16)). Among those in the overweight-to-obese trajectory, the aOR (95 %CI) was 1.45 (0.87-2.43), and thus in the direction of an increased ovarian cancer risk compared to those who maintained a normal weight. CONCLUSION Our findings underscore the need for further research to clarify the role of body fatness across the lifetime in the etiology of ovarian cancer.
Collapse
Affiliation(s)
- Kevin L'Espérance
- Université de Montréal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada; Department of Social and Preventive Medicine, Université de Montréal, Montreal, Quebec, Canada; Department of Urology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Sreenath Madathil
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Jennifer A Ritonja
- Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; St. Mary's Research Centre, Montreal, Quebec, Canada
| | - Michal Abrahamowicz
- Department of Epidemiology, Biostatistics and Occupational Health McGill University, Montreal, Quebec, Canada
| | - Vikki Ho
- Université de Montréal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada; Department of Social and Preventive Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Belinda Nicolau
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health McGill University, Montreal, Quebec, Canada
| | - Jennifer O'Loughlin
- Université de Montréal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada; Department of Social and Preventive Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Anita Koushik
- Université de Montréal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada; Department of Social and Preventive Medicine, Université de Montréal, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; St. Mary's Research Centre, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Tandaric L, Auranen A, Kleinmanns K, DePont Christensen R, Vestrheim Thomsen LC, Wogsland CE, McCormack E, Mäenpää J, Madsen K, Stampe Petersson K, Mirza MR, Bjørge L. Peripheral blood leukocyte signatures as biomarkers in relapsed ovarian cancer patients receiving combined anti-CD73/anti-PD-L1 immunotherapy in arm A of the NSGO-OV-UMB1/ENGOT-OV30 trial. Mol Oncol 2025; 19:1436-1451. [PMID: 39887612 PMCID: PMC12077279 DOI: 10.1002/1878-0261.13811] [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: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025] Open
Abstract
Immune checkpoint inhibitors have demonstrated limited efficacy in overcoming immunosuppression in patients with epithelial ovarian cancer (EOC). Although certain patients experience long-term treatment benefit, reliable biomarkers for responder pre-selection and the distinction of dominant immunosuppressive mechanisms have yet to be identified. Here, we used a 40-marker suspension mass cytometry panel to comprehensively phenotype peripheral blood leukocytes sampled over time from patients with relapsed EOC who underwent combination oleclumab (anti-CD73) and durvalumab (anti-PD-L1) immunotherapy in the NSGO-OV-UMB1/ENGOT-OV30 trial. We found that survival duration was impacted by baseline abundances of total peripheral blood mononuclear cells. Longitudinal analyses revealed a significant increase in CD14+CD16- myeloid cells during treatment, with significant expansion of monocytic myeloid-derived suppressor cells occurring in patients with shorter progression-free survival, who additionally showed a continuous decrease in central memory T-cell abundances. All patients demonstrated significant PD-L1 upregulation over time on most T-cell subsets. Higher CD73 and IDO1 expression on certain leukocytes at baseline significantly positively correlated with longer progression-free survival. Overall, our study proposes potential biomarkers for EOC immunotherapy personalization and response monitoring; however, further validation in larger studies is needed.
Collapse
Affiliation(s)
- Luka Tandaric
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
- Department of Obstetrics and GynecologyHaukeland University HospitalBergenNorway
| | - Annika Auranen
- Department of Obstetrics and Gynecology and Tays Cancer CentreTampere University HospitalFinland
- Nordic Society of Gynaecological Oncology – Clinical Trial Unit (NSGO‐CTU)TampereFinland
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
| | | | - Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
- Department of Obstetrics and GynecologyHaukeland University HospitalBergenNorway
- Department of Health Registry Research and DevelopmentNorwegian Institute of Public HealthOsloNorway
| | - Cara Ellen Wogsland
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
- Kinn Therapeutics ASBergenNorway
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
- Centre for Pharmacy, Department of Clinical ScienceUniversity of BergenNorway
- Department of Internal Medicine, Hematology SectionHaukeland University HospitalBergenNorway
| | - Johanna Mäenpää
- Department of Obstetrics and Gynecology and Tays Cancer CentreTampere University HospitalFinland
- Nordic Society of Gynaecological Oncology – Clinical Trial Unit (NSGO‐CTU)TampereFinland
- Faculty of Medicine and Health TechnologyTampere UniversityFinland
| | - Kristine Madsen
- Nordic Society of Gynaecological Oncology – Clinical Trial Unit (NSGO‐CTU)CopenhagenDenmark
| | - Karen Stampe Petersson
- Nordic Society of Gynaecological Oncology – Clinical Trial Unit (NSGO‐CTU)CopenhagenDenmark
| | - Mansoor Raza Mirza
- Nordic Society of Gynaecological Oncology – Clinical Trial Unit (NSGO‐CTU)CopenhagenDenmark
- Department of OncologyRigshospitalet, Copenhagen University HospitalDenmark
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical ScienceUniversity of BergenNorway
- Department of Obstetrics and GynecologyHaukeland University HospitalBergenNorway
| |
Collapse
|
4
|
Wagner V, Morton M, Dorayappan KDP, Gonzalez A, Yu L, Sakaue T, Conrads T, Maxwell GL, Cosgrove C, Backes F, Wang QE, Cohn DE, O'Malley DM, Selvendiran K. Circulating extracellular vesicles protein expression for early prediction of platinum-resistance in high-grade serous ovarian cancer. Oncogene 2025; 44:1197-1203. [PMID: 40210758 PMCID: PMC12015105 DOI: 10.1038/s41388-025-03382-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025]
Abstract
Platinum resistance in high-grade serous ovarian carcinoma (HGSOC) portends a poor prognosis. Although initial platinum-based chemotherapy response rates are high, 15-20% of patients demonstrate primary resistance to platinum therapy and almost all patients will develop platinum resistance in the recurrent setting. No predictive or diagnostic biomarkers have been utilized specific to platinum resistance. This study aimed to identify candidate biomarkers for platinum resistance in HGSOC using an extracellular vesicle (EV) based approach. We found differentially expressed and distinct EV proteins, namely TMEM205 and CFH, in patients with platinum-resistant (PR) HGSOC compared to those of platinum-sensitive (PS) patients, utilizing liquid chromatography-tandem mass spectrometry (LC-MS/MS). Expression of these EV proteins were validated in patient-derived PR cell lines as well as in clinically relevant mouse models of HGSOC post-platinum therapy. We corroborated these findings using serum samples from patients with PS and PR-HGSOC. Both EV CFH and EV TMEM205 exhibited excellent diagnostic capability for PR as noted by receiver operating characteristic curves with area under the curve values of 0.95 and 0.84, respectively. The high diagnostic performance of TMEM205 and CFH within EVs compared to the relatively poor performance of conventional serum proteins such as Ca125 suggests their robust potential as non-invasive biomarkers for detecting platinum resistance in HGSOC. Furthermore, the ROC curve for the combined biomarker demonstrated excellent diagnostic performance, with an AUC of 0.973, a true positive rate (TPR) of 0.938, and a false positive rate (FPR) of 0.062. Incorporating this multi-protein biomarker panel alongside established biomarkers further enhances diagnostic accuracy. Serum EV CFH and TMEM205 are promising biomarkers for early detection of platinum resistance in HGSOC and may highlight underlying chemoresistance mechanisms, offering potential future therapeutic targets.
Collapse
Affiliation(s)
- Vincent Wagner
- Division of Gynecologic Oncology, Holden Comprehensive Cancer Center, The University of Iowa Health Care, Iowa City, IA, USA
| | - Molly Morton
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | - Anna Gonzalez
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Thomas Conrads
- Inova Women's Service Line and the Inova Schar Cancer Institute, Falls Church, VA, USA
| | - G Larry Maxwell
- Inova Women's Service Line and the Inova Schar Cancer Institute, Falls Church, VA, USA
| | - Casey Cosgrove
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Floor Backes
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Qi-En Wang
- Department of Radiation Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - David E Cohn
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - David M O'Malley
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Xu C, Chen J, Tan M, Tan Q. The role of macrophage polarization in ovarian cancer: from molecular mechanism to therapeutic potentials. Front Immunol 2025; 16:1543096. [PMID: 40330466 PMCID: PMC12052780 DOI: 10.3389/fimmu.2025.1543096] [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: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Ovarian cancer (OC) remains the most lethal gynecological malignancy, primarily due to its late-stage diagnosis, frequent recurrence, and resistance to conventional chemotherapy. A critical factor contributing to OC's aggressiveness is the tumor microenvironment (TME), particularly the presence and polarization of tumor-associated macrophages (TAMs). TAMs, often skewed toward an immunosuppressive M2-like phenotype, facilitate tumor growth, angiogenesis, metastasis, and resistance to therapy. This comprehensive review delves into the multifaceted regulation of macrophage polarization in OC, highlighting key molecular pathways such as PTEN loss, Wnt/β-catenin signaling, NF-κB, Myc, STAT3, and JNK, among others. Additionally, it explores the role of chemokines, non-coding RNAs, and various proteins in modulating TAM phenotypes. Emerging evidence underscores the significance of extracellular vesicles (EVs) and ovarian cancer stem cells (CSCs) in promoting M2 polarization, thereby enhancing tumor progression and therapy resistance. The review also identifies critical biomarkers associated with macrophage polarization, including CD163, LILRB1, MUC2, and others, which hold prognostic and therapeutic potential. Therapeutic strategies targeting TAMs are extensively discussed, encompassing oncolytic viruses, engineered EVs, immunotherapies, nanoparticles, targeted therapies, and natural products. These approaches aim to reprogram TAMs from a pro-tumorigenic M2 state to an anti-tumorigenic M1 phenotype, thereby enhancing immune responses and overcoming resistance to treatments such as chemotherapy and immune checkpoint inhibitors. Furthermore, the review addresses the interplay between macrophage polarization and therapy resistance, emphasizing the need for novel interventions to modulate the TME effectively. By synthesizing current knowledge on macrophage polarization in ovarian cancer, this study underscores the potential of targeting TAMs to improve clinical outcomes and personalize treatment strategies for OC patients. Continued research in this domain is essential to develop robust therapeutic frameworks that can mitigate the immunosuppressive TME and enhance the efficacy of existing and novel cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Qingqing Tan
- Department of Gynecology and Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
6
|
Karimnia N, Wilson AL, Doran BR, Do J, Matthews A, Ho GY, Plebanski M, Jobling TW, Stephens AN, Bilandzic M. A Novel 3D High-Throughput Phenotypic Drug Screening Pipeline to Identify Drugs with Repurposing Potential for the Treatment of Ovarian Cancer. Adv Healthc Mater 2025; 14:e2404117. [PMID: 40109101 PMCID: PMC12023816 DOI: 10.1002/adhm.202404117] [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: 10/22/2024] [Revised: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Ovarian cancer (OC) poses a significant clinical challenge due to its high recurrence rates and resistance to standard therapies, particularly in advanced stages where recurrence is common, and treatment is predominantly palliative. Personalized treatments, while effective in other cancers, remain underutilized in OC due to a lack of reliable biomarkers predicting clinical outcomes. Accordingly, precision medicine approaches are limited, with PARP inhibitors showing efficacy only in specific genetic contexts. Drug repurposing offers a promising, rapidly translatable strategy by leveraging existing pharmacological data to identify new treatments for OC. Patient-derived polyclonal spheroids, isolated from ascites fluid closely mimic the clinical behavior of OC, providing a valuable model for drug testing. Using these spheroids, a high-throughput drug screening pipeline capable of evaluating both cytotoxicity and anti-migratory properties of a diverse drug library, including FDA-approved, investigational, and newly approved compounds is developed. The findings highlight the importance of 3D culture systems, revealing a poor correlation between drug efficacy in traditional 2D models and more clinically relevant 3D spheroids. This approach has expedited the identification of promising candidates, such as rapamycin, which demonstrated limited activity as a monotherapy but synergized effectively with standard treatments like cisplatin and paclitaxel in vitro. In combination with platinum-based therapy, Rapamycin led to significant in vitro cytotoxicity and a marked reduction in tumor burden in a syngeneic in vivo model. This proof-of-concept study underscores the potential of drug repurposing to rapidly advance new treatments into clinical trials for OC, offering renewed hope for patients with advanced disease.
Collapse
Affiliation(s)
- Nazanin Karimnia
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Brittany R. Doran
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Jennie Do
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amelia Matthews
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Gwo Y. Ho
- School of Clinical SciencesMonash University27 Rainforest WalkClaytonVIC3168Australia
- Department of OncologyMonash Health823–865 Centre RdBentleighVIC3165Australia
| | - Magdalena Plebanski
- School of Health and Biomedical SciencesRMIT UniversityBundoora campus West203, 1/2 Plenty RdBundooraVIC3083Australia
| | - Thomas W. Jobling
- Monash Medical CentreDepartment of Gynecology OncologyMonash Health823–865 Centre RdBentleigh EastVIC3165Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| |
Collapse
|
7
|
Zhang XY, Zhao CY, Dong JM, Du CP, Zhang CL, Yang AJ, Zhou Q, Liu W, Dang Y, Shang LN, Zhou YN, Wang YP, Wang CY, Wang M, Li M. ADAM9 mediates Cisplatin resistance in gastric cancer cells through DNA damage response pathway. Med Oncol 2025; 42:122. [PMID: 40108033 DOI: 10.1007/s12032-025-02645-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Gastric cancer is one of the most common malignant tumors in the world. The occurrence of chemotherapy resistance seriously affects the survival and prognosis of middle and advanced patients. Enhancing DNA repair ability is one of the important mechanisms of chemotherapy resistance. ADAM9, a member of the disintegrin and metalloproteinase family, is involved in many biological processes, such as tumor cells proliferation, apoptosis, invasion and migration, vascular invasion, and drug resistance. In this study, we found that the high expression of ADAM9 in gastric cancer tissues was associated with a variety of clinicopathological factors and poor prognosis in patients. Gastric cancer cells with high ADAM9 expression reduced sensitivity to Cisplatin, decreased DNA damage, increased expression of ATM and CHK2, the key proteins in DNA damage repair pathway, and improved cancer cells survival rate. Further studies showed that the expression of ADAM9 was selectively interfered with gastric cancer cells, the expression levels of ATM and CHK2 were decreased, while the expression of damage protein γ-H2AX was significantly increased, the degree of DNA damage was increased, and the sensitivity of gastric cancer cells to Cisplatin was significantly enhanced. It is suggested that ADAM9 is involved in Cisplatin resistance in gastric cancer cells, and its mechanism is related to the activation of ATM-CHK2 pathway in DNA damage repair. These data demonstrate that ADAM9 plays a pro-cancer role and mediates Cisplatin resistance in gastric cancer, which may be a new target to overcome chemotherapy resistance.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chan-Yuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Ming Dong
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Cun-Pu Du
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chen-Li Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Ai-Jun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Yun Dang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Gansu Provincial Maternity and Child-Care Hospital/Gansu Provincial Central Hospital, Lanzhou, China
| | - Li-Na Shang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Yong-Ning Zhou
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yu-Ping Wang
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Chen-Yu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China.
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China.
- Experimental Teaching Center of Basic Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Wang J, Zhu W, Li X, Wu Y, Ma W, Wang Y, Zhao W, Wei F, Wang W. Transcriptome analysis of ovarian cancer uncovers association between tumor-related inflammation/immunity and patient outcome. Front Pharmacol 2025; 16:1500251. [PMID: 39981173 PMCID: PMC11839622 DOI: 10.3389/fphar.2025.1500251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Background Epithelial ovarian cancer (EOC) is a cancer that affects the female reproductive system and is highly lethal. It poses significant challenges in terms of treatment and often has a poor prognosis. In recent years, with the advent of PARPi, the treatment of ovarian cancer has entered a new stage of full-process management. Although more and more drugs have been approved, the therapeutic effect of PARPi is still very limited. With the rapid development of PD-1/PD-L1, CTLA-4, oncolytic viruses, cancer vaccines, adoptive cell therapy, etc., tumor immunotherapy has provided new opportunities for the treatment of ovarian cancer. Methods This study used comprehensive transcriptome analysis across multiple databases to gather gene transcripts and clinical features of normal ovarian samples and tissue samples from ovarian cancer. The aim was to explore the mechanisms underlying tumor immunotherapy resistance and to reveal the relationship between ovarian cancer's immune microenvironment and genes linked to inflammation. Various R packages were used for differential gene analysis, enrichment analysis, co-expression network construction, and prognostic model building. Results It has been found that the prognosis of ovarian cancer patients is closely associated with sets of genes involved in inflammation. The immune infiltration microenvironment, clinicopathological features, and survival rates differed significantly between two inflammatory gene expression patterns identified using cluster and immune microenvironment analyses. Further analysis revealed that the high-risk group had a higher abundance of M2-type macrophage infiltration, more active anti-tumor immune response, higher tumor stemness score, potentially worse prognosis, and lower response rates to multiple chemotherapy drugs and immune checkpoint inhibitors. Conclusion These findings provide new perspectives and potential targets for immunotherapy and prognostic evaluation of ovarian cancer and offer new strategies and directions for clinical treatment and patient management. This study provides crucial information to further our comprehension of drug response mechanisms and tumor immunotherapy. It offers new strategies and methods for the treatment and prognostic improvement of ovarian cancer.
Collapse
Affiliation(s)
- Jingfang Wang
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenrui Zhu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xia Li
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuanyuan Wu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenhui Ma
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yangzhou Wang
- Department of Stomatology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Weihong Zhao
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fang Wei
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenhao Wang
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
9
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [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: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
10
|
Yang LR, Yang M, Chen LL, Shen YL, He Y, Meng ZT, Wang WQ, Li F, Liu ZJ, Li LH, Wang YF, Luo XL. Machine learning for epithelial ovarian cancer platinum resistance recurrence identification using routine clinical data. Front Oncol 2024; 14:1457294. [PMID: 39582538 PMCID: PMC11581972 DOI: 10.3389/fonc.2024.1457294] [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: 06/30/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Background Most epithelial ovarian cancer (EOC) eventually develops recurrence. Identification of high-risk patients can prompt earlier intervention and improve long-term outcomes. We used laboratory and clinical data to create models based on machine learning for EOC platinum resistance recurrence identification. Methods This study was designed as a retrospective cohort analysis. Initially, we identified 1,392 patients diagnosed with epithelial ovarian cancer who underwent platinum-based chemotherapy at Yunnan Cancer Hospital between January 1, 2012, and June 30, 2022. We collected data on the patients' clinicopathologic characteristics, routine laboratory results, surgical information, details of chemotherapy regimens, and survival outcomes. Subsequently, to identify relevant variables influencing the recurrence of platinum resistance, we screened thirty potential factors using two distinct variable selection methods: Lasso regression and multiple logistic regression analysis. Following this screening process, five machine learning algorithms were employed to develop predictive models based on the selected variables. These included decision tree analysis (DTA), K-Nearest neighbor (KNN), support vector machine (SVM), random forest (RF), and eXtreme gradient boosting (XGBoost). The performance of these models was compared against that of traditional logistic regression. To ensure robust internal validation and facilitate comparison among model performance metrics, a five-fold cross-validation method was implemented. Key performance indicators for the models included the area under the receiver operating characteristic curve (AUC), sensitivity, specificity, and average accuracy. Finally, we will visualize these models through nomograms, decision tree diagrams, variable importance plots, etc., to assist clinicians in their practice. Results Multiple logistic regression analysis identified eight variables associated with platinum resistance recurrence. In the lasso regression, seven variables were selected. Based on the findings from both Lasso regression and multiple logistic regression analysis, models were developed using these 7 and 8 factors. Among these, the XGBoost model derived from multiple logistic regression exhibited superior performance and demonstrated good discrimination during internal validation, achieving an AUC of 0.784, a sensitivity of 0.735, a specificity of 0.713, an average accuracy of 80.4%, with a cut-off value set at 0.240. Conversely, the LR model based on lasso regression yielded commendable results as well; it achieved an AUC of 0.738, a sensitivity of 0.541, a specificity of 0.836, with a cut-off value established at 0.154 and an accuracy rate of 79.6%. Finally, we visualized both models through nomograms to illustrate the significance of each variable involved in their development. Conclusions We have successfully developed predictive models for platinum-resistant recurrence of epithelial ovarian cancer, utilizing routine clinical and laboratory data. Among these models, the XGBoost model-derived from variables selected through multiple logistic regression-demonstrated the best performance. It exhibited high AUC values and average accuracy during internal validation, making it a recommended tool for clinical use. However, due to variations in time and context, influencing factors may change over time; thus, continuous evolution of the model is necessary. We propose a framework for this ongoing model adaptation.
Collapse
Affiliation(s)
- Li-Rong Yang
- Hematology Oncology Department, the Southern Central Hospital of Yunnan Province, Honghe, Yunnan, China
| | - Mei Yang
- Geriatric Oncology Department, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Liu-Lin Chen
- Hematology Oncology Department, the Southern Central Hospital of Yunnan Province, Honghe, Yunnan, China
| | - Yong-Lin Shen
- Hematology Oncology Department, the Southern Central Hospital of Yunnan Province, Honghe, Yunnan, China
| | - Yuan He
- Geriatric Oncology Department, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zong-Ting Meng
- Geriatric Oncology Department, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wan-Qi Wang
- Geriatric Oncology Department, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Feng Li
- Department of Oncology, the Pingxiang People’s Hospital, Pingxiang, Jiangxi, China
| | - Zhi-Jin Liu
- Department of Oncology, The First Hospital of Nanchang, Nangchang, Jiangxi, China
| | - Lin-Hui Li
- Hematology Oncology Department, the Southern Central Hospital of Yunnan Province, Honghe, Yunnan, China
| | - Yu-Feng Wang
- Geriatric Oncology Department, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xin-Lei Luo
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| |
Collapse
|
11
|
Overchuk M, Rickard BP, Tulino J, Tan X, Ligler FS, Huang HC, Rizvi I. Overcoming the effects of fluid shear stress in ovarian cancer cell lines: Doxorubicin alone or photodynamic priming to target platinum resistance. Photochem Photobiol 2024; 100:1676-1693. [PMID: 38849970 PMCID: PMC11568959 DOI: 10.1111/php.13967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 06/09/2024]
Abstract
Resistance to platinum-based chemotherapies remains a significant challenge in advanced-stage high-grade serous ovarian carcinoma, and patients with malignant ascites face the poorest outcomes. It is, therefore, important to understand the effects of ascites, including the associated fluid shear stress (FSS), on phenotypic changes and therapy response, specifically FSS-induced chemotherapy resistance and the underlying mechanisms in ovarian cancer. This study investigated the effects of FSS on response to cisplatin, a platinum-based chemotherapy, and doxorubicin, an anthracycline, both of which are commonly used to manage advanced-stage ovarian cancer. Consistent with prior research, OVCAR-3 and Caov-3 cells cultivated under FSS demonstrated significant resistance to cisplatin. Examination of the role of mitochondria revealed an increase in mitochondrial DNA copy number and intracellular ATP content in cultures grown under FSS, suggesting that changes in mitochondria number and metabolic activity may contribute to platinum resistance. Interestingly, no resistance to doxorubicin was observed under FSS, the first such observation of a lack of resistance under these conditions. Finally, this study demonstrated the potential of photodynamic priming using benzoporphyrin derivative, a clinically approved photosensitizer that localizes in part to mitochondria and endoplasmic reticula, to enhance the efficacy of cisplatin, but not doxorubicin, thereby overcoming FSS-induced platinum resistance.
Collapse
Affiliation(s)
- Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Justin Tulino
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Xianming Tan
- Department of Biostatistics, University of North Carolina School of Public Health, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Frances S. Ligler
- Department of Biomedical Engineering, Texas A&M University, Collage Station, TX, 77843 USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Perez-Fidalgo JA, Gálvez-Montosa F, Guerra EM, Madariaga A, Manzano A, Martin-Lorente C, Rubio-Pérez MJ, Alarcón J, Barretina-Ginesta MP, Gaba L. SEOM-GEICO clinical guideline on epithelial ovarian cancer (2023). Clin Transl Oncol 2024; 26:2758-2770. [PMID: 39008159 PMCID: PMC11467069 DOI: 10.1007/s12094-024-03531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/16/2024]
Abstract
In recent years, the incorporation of new strategies to the therapeutic armamentarium has completely changed the outcomes of epithelial ovarian cancer (EOC). The identification of new predictive and prognostic biomarkers has also enabled the selection of those patients more likely to respond to targeted agents. Nevertheless, EOC is still a highly lethal disease and resistance to many of these new agents is common. The objective of this guideline is to summarize the most relevant strategies to manage EOC, to help the clinician throughout the challenging diagnostic and therapeutic processes and to provide evidence-based recommendations.
Collapse
Affiliation(s)
| | | | | | - Ainhoa Madariaga
- Department of Medical Oncology, 12 de Octubre University Hospital, Madrid, Spain
| | | | | | | | | | - María Pilar Barretina-Ginesta
- Institut Català d'Oncologia, Medical Oncology Department, Precision Oncology Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Medical Sciences Department, Universitat de Girona, Girona, Spain
| | - Lydia Gaba
- Medical Oncology Department, Hospital Clinic, Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Garbuzenko OB, Sapiezynski J, Girda E, Rodriguez-Rodriguez L, Minko T. Personalized Versus Precision Nanomedicine for Treatment of Ovarian Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307462. [PMID: 38342698 PMCID: PMC11316847 DOI: 10.1002/smll.202307462] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
The response to treatment is substantially varied between individual patients with ovarian cancer. However, chemotherapy treatment plans rarely pay sufficient attention to the mentioned factors. Instead, standardized treatment protocols are usually employed for most ovarian cancer patients. Variations in an individual's sensitivity to drugs significantly limit the effectiveness of treatment in some patients and lead to severe toxicities in others. In the present investigation, a nanotechnology-based approach for personalized treatment of ovarian carcinoma (the most lethal type of gynecological cancer) constructed on the individual genetic profile of the patient's tumor is developed and validated. The expression of predefined genes and proteins is analyzed for each patient sample. Finally, a mixture of the complex nanocarrier-based targeted delivery system containing drug(s)/siRNA(s)/targeted peptide is selected from the pre-synthesized bank and tested in vivo on murine cancer model using cancer cells isolated from tumors of each patient. Based on the results of the present study, an innovative approach and protocol for personalized treatment of ovarian cancer are suggested and evaluated. The results of the present study clearly show the advantages and perspectives of the proposed individual treatment approach.
Collapse
Affiliation(s)
- Olga B. Garbuzenko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
| | - Justin Sapiezynski
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
| | - Eugenia Girda
- Department of Gynecology Oncology, Robert Wood Johnson School of Medicine, Rutgers the State University of New Jersey, New Brunswick, NJ USA 08901
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA 08901
| | - Lorna Rodriguez-Rodriguez
- Department of Surgery, Division of Gynecologic Oncology, City of Hope National Medical Center, Duarte, CA 91010
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers the State University of New Jersey, Piscataway, NJ USA 08854
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA 08901
| |
Collapse
|
14
|
Dinkins K, Barton W, Wheeler L, Smith HJ, Mythreye K, Arend RC. Targeted therapy in high grade serous ovarian Cancer: A literature review. Gynecol Oncol Rep 2024; 54:101450. [PMID: 39092168 PMCID: PMC11292514 DOI: 10.1016/j.gore.2024.101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
Ovarian cancer continues to have a high mortality rate despite therapeutic advances. Traditionally, treatment has focused on surgery followed by systemic platinum- based chemotherapy. Unfortunately, most patients develop resistance to platinum agents, highlighting the need for targeted therapies. PARP inhibitors and anti-angiogenic agents, such as bevacizumab, have more recently changed upfront therapy. Unfortunately, other targeted therapies including immunotherapy have not seen the same success. Emerging therapeutic targets and modalities such as small molecule tyrosine kinase inhibitors, lipid metabolism targeting agents, gene therapy, ribosome targeted drugs as well as several other therapeutic classes have been and are currently under investigation. In this review, we discuss targeted therapies in high grade serous ovarian cancer from preclinical studies to phase III clinical trials.
Collapse
Affiliation(s)
- Kaitlyn Dinkins
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Wade Barton
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Lauren Wheeler
- Lister Hill Library, University of Alabama at Birmingham, Birmingham, AL
| | - Haller J. Smith
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Karthikeyan Mythreye
- Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Rebecca C. Arend
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
15
|
Xing L, Wang Z, Feng Y, Luo H, Dai G, Sang L, Zhang C, Qian J. The biological roles of CD47 in ovarian cancer progression. Cancer Immunol Immunother 2024; 73:145. [PMID: 38832992 PMCID: PMC11150368 DOI: 10.1007/s00262-024-03708-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
Ovarian cancer is one of the most lethal malignant tumors, characterized by high incidence and poor prognosis. Patients relapse occurred in 65-80% after initial treatment. To date, no effective treatment has been established for these patients. Recently, CD47 has been considered as a promising immunotherapy target. In this paper, we reviewed the biological roles of CD47 in ovarian cancer and summarized the related mechanisms. For most types of cancers, the CD47/Sirpα immune checkpoint has attracted the most attention in immunotherapy. Notably, CD47 monoclonal antibodies and related molecules are promising in the immunotherapy of ovarian cancer, and further research is needed. In the future, new immunotherapy regimens targeting CD47 can be applied to the clinical treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Linan Xing
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Zhao Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Yue Feng
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Haixia Luo
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Guijiang Dai
- Department of Comprehensive Office, The Second Affiliated Hospital of MuDanjiang Medical University, Mudanjiang, 157009, People's Republic of China
| | - Lin Sang
- Department of Obstetrics and Gynecology, People's Hospital of Anji, Huzhou, 310022, People's Republic of China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
16
|
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: 4] [Impact Index Per Article: 4.0] [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
|
17
|
Nie X, Xu T, Zhang L, Cheng W. The Gustave Roussy immune score as a novel scoring system for predicting platinum resistance in advanced high-grade serous ovarian cancer. Eur J Obstet Gynecol Reprod Biol 2024; 294:97-104. [PMID: 38219610 DOI: 10.1016/j.ejogrb.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/16/2023] [Accepted: 01/03/2024] [Indexed: 01/16/2024]
Abstract
OBJECTIVE This study was designed to investigate the relationship between the Gustave-Roussy immune score (GRIm-score) and platinum resistance in patients with advanced high-grade serous ovarian cancer (HGSOC). METHODS We conducted a retrospective study of patients diagnosed with advanced HGSOC between January 2017 and December 2020. A nomogram was developed to predict the risk of platinum resistance. Receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA) were used to validate the nomogram. Bootstrap analysis was utilized for internal validation. Additionally, we analyzed the risk factors for platinum resistance in patients who received neoadjuvant chemotherapy (NACT). RESULTS A total of 232 patients with advanced HGSOC were included, 52 (22.4 %) of whom experienced relapse with platinum resistance. Multivariate logistic regression analysis revealed that high GRIm-score (OR = 4.174, P < 0.001), NACT (OR = 2.706, P = 0.017), PLT > 260 (OR = 2.233, P = 0.037) and non-R0 (OR = 2.526, P = 0.012) were independent risk factors for platinum resistance. The area under the curve (AUC) of the model was 0.802 (95 % CI 0.736-0.868), and the internally validated AUC of 1000 bootstrap samples was 0.798 (95 % CI 0.725-0.862). In NACT-treated patients, univariate and multivariate logistic regression analyses revealed that a low KELIM score (OR = 10.405, P = 0.001) and PLT > 260 (OR = 4.611, P = 0.014) were independent risk factors for platinum resistance. CONCLUSION The GRIm-score and PLT count are important prognostic factors in patients with HGSOC. For precision treatment, the status of partially platinum-sensitive patients should also be considered.
Collapse
Affiliation(s)
- Xianglin Nie
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Ting Xu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Lin Zhang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
18
|
Li W, Zhang K, Wang W, Liu Y, Huang J, Zheng M, Li L, Zhang X, Xu M, Chen G, Wang L, Zhang S. Combined inhibition of HER2 and VEGFR synergistically improves therapeutic efficacy via PI3K-AKT pathway in advanced ovarian cancer. J Exp Clin Cancer Res 2024; 43:56. [PMID: 38403634 PMCID: PMC10895844 DOI: 10.1186/s13046-024-02981-5] [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: 09/16/2023] [Accepted: 02/11/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a prevalent malignancy in the female reproductive system, and developing effective targeted therapies for this disease remains challenging. The aim of this study was to use clinically-relevant OC models to evaluate the therapeutic effectiveness of RC48, an antibody-drug conjugate (ADC) targeting HER2, either alone or in combination with the VEGFR inhibitor Cediranib Maleate (CM), for the treatment of advanced OC. METHODS OC tumor specimens and cell lines were analyzed to determine HER2 and VEGFR expression by Western blot, immunocytochemistry and immunofluorescence. Moreover, the OC cell lines, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models were treated with RC48 and/or CM and then subjected to cell proliferation, viability, apoptosis, and tumor growth analyses to evaluate the feasibility of combination therapy for OC both in vitro and in vivo. Additionally, RNA-Seq was performed to investigate the critical mechanism underlying the combination therapy of RC48 and CM. RESULTS Our results demonstrated that RC48 alone effectively targeted and inhibited the growth of HER2-positive OC tumors in both cell lines and PDX models. Furthermore, the combination of RC48 and CM synergistically induced tumor regression in human OC cell lines, as well as CDX and PDX models. Mechanistically, we observed that the combination treatment inhibited the growth of OC cells involved inducing apoptosis and suppressing cell motility. RNA-seq analysis provided further mechanistic insights and revealed that co-administration of RC48 and CM downregulated multiple cancer-related pathways, including the AKT/mTOR pathway, cell cycle, and cell proliferation. Notably, our data further confirmed that the PI3K-AKT pathway played a key role in the inhibition of proliferation triggered by combinational treatment of RC48 and CM in OC cells. CONCLUSIONS These findings provide a preclinical framework supporting the potential of dual targeting HER2 and VEGFR as a promising therapeutic strategy to improve outcomes in patients with OC.
Collapse
Affiliation(s)
- Weisong Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Kai Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Wenjun Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Liu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China
| | - Jianming Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Meihong Zheng
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Ling Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Xinyu Zhang
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Minjuan Xu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China.
| | - Guofang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, 200092, China.
| | - Liefeng Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| | - Shuyong Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
19
|
Tavares V, Marques IS, Melo IGD, Assis J, Pereira D, Medeiros R. Paradigm Shift: A Comprehensive Review of Ovarian Cancer Management in an Era of Advancements. Int J Mol Sci 2024; 25:1845. [PMID: 38339123 PMCID: PMC10856127 DOI: 10.3390/ijms25031845] [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: 12/31/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Ovarian cancer (OC) is the female genital malignancy with the highest lethality. Patients present a poor prognosis mainly due to the late clinical presentation allied with the common acquisition of chemoresistance and a high rate of tumour recurrence. Effective screening, accurate diagnosis, and personalised multidisciplinary treatments are crucial for improving patients' survival and quality of life. This comprehensive narrative review aims to describe the current knowledge on the aetiology, prevention, diagnosis, and treatment of OC, highlighting the latest significant advancements and future directions. Traditionally, OC treatment involves the combination of cytoreductive surgery and platinum-based chemotherapy. Although more therapeutical approaches have been developed, the lack of established predictive biomarkers to guide disease management has led to only marginal improvements in progression-free survival (PFS) while patients face an increasing level of toxicity. Fortunately, because of a better overall understanding of ovarian tumourigenesis and advancements in the disease's (epi)genetic and molecular profiling, a paradigm shift has emerged with the identification of new disease biomarkers and the proposal of targeted therapeutic approaches to postpone disease recurrence and decrease side effects, while increasing patients' survival. Despite this progress, several challenges in disease management, including disease heterogeneity and drug resistance, still need to be overcome.
Collapse
Affiliation(s)
- Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology of Porto (IPOP), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-072 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
20
|
Tadić V, Zhang W, Brozovic A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim Biophys Acta Rev Cancer 2024; 1879:189052. [PMID: 38097143 DOI: 10.1016/j.bbcan.2023.189052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive type of epithelial ovarian cancer, with high recurrence rate and chemoresistance being the main issues in its clinical management. HGSOC is specifically challenging due to the metastatic dissemination via spheroids in the ascitic fluid. The HGSOC spheroids represent the invasive and chemoresistant cellular fraction, which is impossible to investigate in conventional two-dimensional (2D) monolayer cell cultures lacking critical cell-to-cell and cell-extracellular matrix interactions. Three-dimensional (3D) HGSOC cultures, where cells aggregate and exhibit relevant interactions, offer a promising in vitro model of peritoneal metastasis and multicellular drug resistance. This review summarizes recent studies of HGSOC in 3D culture conditions and highlights the role of multicellular HGSOC spheroids and ascitic environment in HGSOC metastasis and chemoresistance.
Collapse
Affiliation(s)
- Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia
| | - Wei Zhang
- Department of Engineering Mechanics, Dalian University of Technology, Linggong Road 2, Dalian CN-116024, China
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia.
| |
Collapse
|
21
|
Bachmann C. New Achievements from Molecular Biology and Treatment Options for Refractory/Relapsed Ovarian Cancer-A Systematic Review. Cancers (Basel) 2023; 15:5356. [PMID: 38001616 PMCID: PMC10669965 DOI: 10.3390/cancers15225356] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Ovarian cancer (OC) has a high rate of mortality and is the fifth most common cause of death in females all over the world. The etiology is still unclear. Numerous factors such as smoking, obesity, and unhealthy diet may affect the risk of OC. Having a family history of breast and OC is one of the main risks for developing OC. Mutations of BRCA1/2 are associated with OC risk as well. The histopathological classification of OC reveals the four most common types: serous, clear cell, endometrioid, and mucinous; these are epithelial OC types, and other types are rare. Furthermore, OC can be subdivided into types I and II. Type I tumors are most probably caused by atypical proliferative tumors. Type II tumors include high-grade carcinoma of the serous type, carcinosarcoma, and carcinoma, which are not differentiated and generally originate from tubal intraepithelial carcinoma of the serous type. Typically, type I tumors are present in early stages, usually with good prognosis. Type II tumors are classified as high-grade tumors and are most often diagnosed at advanced FIGO stages with poor prognosis. High-grade serous OC accounts for 90% of serous OC. Tumor heterogeneity aggravates OC treatment. The standard care for primary epithelial ovarian cancer (EOC) is cytoreductive surgery followed by platinum-based chemotherapy. Neoadjuvant chemotherapy can be used in certain cases followed by cytoreductive surgery. The main prognostic factor is complete tumor resection. However, about 70% of patients relapse. Resistance to chemotherapeutic agents remains a major challenge in EOC treatment, in which many different factors are involved. In recent years, the examination of molecular parameters and their prognostic impact has become increasingly relevant in EOC, and furthermore, the use of immunotherapy has expanded the therapeutic range. As the clinical need is greatest for relapsed patients, this systematic review will focus on recent advances in molecular biology with prognostic and predictive markers and treatment options for recurrent/refractory OC. Inclusion criteria for the review: potential prospective or predictive biomarkers in preclinical or clinical use in relapsed and refractory OC, prognostic impact, clinical and preclinical trials, and immunotherapy. Exclusion criteria for the review: primary OC, no full text or abstract available, not the topic mentioned above, and text not available in English. Risk of bias: the included studies were evaluated descriptively for the topics mentioned above, and data were not compared with each other. The objective is to highlight the molecular mechanisms of the most promising targeted agents under clinical investigation to demonstrate their potential relevance in recurrent/refractory OC.
Collapse
Affiliation(s)
- Cornelia Bachmann
- Department of Womens' Health, University Tübingen, 72070 Tübingen, Germany
| |
Collapse
|
22
|
Ashoori F, Hajipour-Verdom B, Satari M, Abdolmaleki P. Polyethylenimine-based iron oxide nanoparticles enhance cisplatin toxicity in ovarian cancer cells in the presence of a static magnetic field. Front Oncol 2023; 13:1217800. [PMID: 37771439 PMCID: PMC10522916 DOI: 10.3389/fonc.2023.1217800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Background Drug resistance in cancer cells is a major concern in chemotherapy. Cisplatin (CIS) is one of the most effective chemotherapeutics for ovarian cancer. Here, we investigated an experimental approach to increase CIS cytotoxicity and overcome cell resistance using nanoparticle-based combination treatments. Methods Polyethylenimine (PEI)-based magnetic iron oxide nanocomplexes were used for drug delivery in genetically matched CIS-resistant (A2780/CP) and -sensitive (A2780) ovarian cancer cells in the presence of a 20 mT static magnetic field. Magnetic nanoparticles (MNPs) were synthesized and bonded to PEI cationic polymers to form binary complexes (PM). The binding of CIS to the PM binary complexes resulted in the formation of ternary complexes PM/C (PEI-MNP/CIS) and PMC (PEI-MNP-CIS). Results CIS cytotoxicity increased at different concentrations of CIS and PEI in all binary and ternary delivery systems over time. Additionally, CIS induced cell cycle arrest in the S and G2/M phases and reactive oxygen species production in both cell lines. Ternary complexes were more effective than binary complexes at promoting apoptosis in the treated cells. Conclusion PEI-based magnetic nanocomplexes can be considered novel carriers for increasing CIS cytotoxicity and likely overcoming drug resistance of ovarian cancer cells.
Collapse
Affiliation(s)
- Faranak Ashoori
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behnam Hajipour-Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Satari
- Department of Biology, Faculty of Sciences, Malayer University, Malayer, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
23
|
Li H, Cai S, Deng L, Xiao Z, Guo Q, Qiang J, Gong J, Gu Y, Liu Z. Prediction of platinum resistance for advanced high-grade serous ovarian carcinoma using MRI-based radiomics nomogram. Eur Radiol 2023; 33:5298-5308. [PMID: 36995415 DOI: 10.1007/s00330-023-09552-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVE This study aimed to explore the value of a radiomics nomogram to identify platinum resistance and predict the progression-free survival (PFS) of patients with advanced high-grade serous ovarian carcinoma (HGSOC). MATERIALS AND METHODS In this multicenter retrospective study, 301 patients with advanced HGSOC underwent radiomics features extraction from the whole primary tumor on contrast-enhanced T1WI and T2WI. The radiomics features were selected by the support vector machine-based recursive feature elimination method, and then the radiomics signature was generated. Furthermore, a radiomics nomogram was developed using the radiomics signature and clinical characteristics by multivariable logistic regression. The predictive performance was evaluated using receiver operating characteristic analysis. The net reclassification index (NRI), integrated discrimination improvement (IDI), and decision curve analysis (DCA) were used to compare the clinical utility and benefits of different models. RESULTS Five features significantly correlated with platinum resistance were selected to construct the radiomics model. The radiomics nomogram, combining radiomics signatures with three clinical characteristics (FIGO stage, CA-125, and residual tumor), had a higher area under the curve (AUC) compared with the clinical model alone (AUC: 0.799 vs 0.747), with positive NRI and IDI. The net benefit of the radiomics nomogram is typically higher than clinical-only and radiomics-only models. Kaplan-Meier survival analysis showed that the radiomics nomogram-defined high-risk groups had shorter PFS compared with the low-risk groups in patients with advanced HGSOC. CONCLUSIONS The radiomics nomogram can identify platinum resistance and predict PFS. It helps make the personalized management of advanced HGSOC. KEY POINTS • The radiomics-based approach has the potential to identify platinum resistance and can help make the personalized management of advanced HGSOC. • The radiomics-clinical nomogram showed improved performance compared with either of them alone for predicting platinum-resistant HGSOC. • The proposed nomogram performed well in predicting the PFS time of patients with low-risk and high-risk HGSOC in both training and testing cohorts.
Collapse
Affiliation(s)
- Haiming Li
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangzhou, 510080, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Songqi Cai
- Department of Radiology, Zhongshan Hospital, FudanUniversity, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Department of Cancer Center, Zhongshan Hospital, FudanUniversity, Shanghai, 200032, China
| | - Lin Deng
- Department of Radiology, Jinshan Hospital, FudanUniversity, Shanghai, 201508, China
| | - Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qinhao Guo
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, FudanUniversity, Shanghai, 201508, China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
24
|
Yang S, Fei W, Zhao Y, Wang F, Ye Y, Wang F. Combat Against Gynecological Cancers with Blood Vessels as Entry Point: Anti-Angiogenic Drugs, Clinical Trials and Pre-Clinical Nano-Delivery Platforms. Int J Nanomedicine 2023; 18:3035-3046. [PMID: 37312935 PMCID: PMC10259534 DOI: 10.2147/ijn.s411761] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Angiogenesis is an essential mechanism for the progression of gynecological cancers. Although approved anti-angiogenic drugs have demonstrated clinical efficacy in treating gynecological cancers, the full potential of therapeutic strategies based on tumor blood vessels has not yet been realized. This review summarizes the latest angiogenesis mechanisms involved in the progression of gynecological cancers and discusses the current clinical practice of approved anti-angiogenic drugs and related clinical trials. Given the close relationship between gynecological cancers and blood vessels, we highlight more delicate strategies for regulating tumor vessels, including wise drug combinations and smart nano-delivery platforms to achieve highly efficient drug delivery and overall vessel microenvironment regulation. We also address current challenges and future opportunities in this field. We aim to generate interest in therapeutic strategies that target blood vessels as a key entry point and offer new potential and inspiration for combating gynecological cancers.
Collapse
Affiliation(s)
- Shan Yang
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| | - Weidong Fei
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| | - Yunchun Zhao
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| | - Fengmei Wang
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| | - Yiqing Ye
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| | - Fenfen Wang
- Department of Gynecology Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, Peoples Republic of China
| |
Collapse
|
25
|
Kacsir I, Sipos A, Major E, Bajusz N, Bényei A, Buglyó P, Somsák L, Kardos G, Bai P, Bokor É. Half-Sandwich Type Platinum-Group Metal Complexes of C-Glucosaminyl Azines: Synthesis and Antineoplastic and Antimicrobial Activities. Molecules 2023; 28:molecules28073058. [PMID: 37049820 PMCID: PMC10096180 DOI: 10.3390/molecules28073058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
While platinum-based compounds such as cisplatin form the backbone of chemotherapy, the use of these compounds is limited by resistance and toxicity, driving the development of novel complexes with cytostatic properties. In this study, we synthesized a set of half-sandwich complexes of platinum-group metal ions (Ru(II), Os(II), Ir(III) and Rh(III)) with an N,N-bidentate ligand comprising a C-glucosaminyl group and a heterocycle, such as pyridine, pyridazine, pyrimidine, pyrazine or quinoline. The sugar-containing ligands themselves are unknown compounds and were obtained by nucleophilic additions of lithiated heterocycles to O-perbenzylated 2-nitro-glucal. Reduction of the adducts and, where necessary, subsequent protecting group manipulations furnished the above C-glucosaminyl heterocycles in their O-perbenzylated, O-perbenzoylated and O-unprotected forms. The derived complexes were tested on A2780 ovarian cancer cells. Pyridine, pyrazine and pyridazine-containing complexes proved to be cytostatic and cytotoxic on A2780 cells, while pyrimidine and quinoline derivatives were inactive. The best complexes contained pyridine as the heterocycle. The metal ion with polyhapto arene/arenyl moiety also impacted on the biological activity of the complexes. Ruthenium complexes with p-cymene and iridium complexes with Cp* had the best performance in ovarian cancer cells, followed by osmium complexes with p-cymene and rhodium complexes with Cp*. Finally, the chemical nature of the protective groups on the hydroxyl groups of the carbohydrate moiety were also key determinants of bioactivity; in particular, O-benzyl groups were superior to O-benzoyl groups. The IC50 values of the complexes were in the low micromolar range, and, importantly, the complexes were less active against primary, untransformed human dermal fibroblasts; however, the anticipated therapeutic window is narrow. The bioactive complexes exerted cytostasis on a set of carcinomas such as cell models of glioblastoma, as well as breast and pancreatic cancers. Furthermore, the same complexes exhibited bacteriostatic properties against multiresistant Gram-positive Staphylococcus aureus and Enterococcus clinical isolates in the low micromolar range.
Collapse
Affiliation(s)
- István Kacsir
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
- Doctoral School of Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
- The Hungarian Academy of Sciences, Center of Excellence, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, H-4032 Debrecen, Hungary
| | - Evelin Major
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Nikolett Bajusz
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Attila Bényei
- Department of Physical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
| | - Péter Buglyó
- Department of Inorganic & Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
- The Hungarian Academy of Sciences, Center of Excellence, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, H-4032 Debrecen, Hungary
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, H-4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: (P.B.); (É.B.); Tel.: +36-524-123-45 (P.B.); +36-525-129-00 (ext. 22474) (É.B.)
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
- Correspondence: (P.B.); (É.B.); Tel.: +36-524-123-45 (P.B.); +36-525-129-00 (ext. 22474) (É.B.)
| |
Collapse
|
26
|
Madariaga A, Garg S, Tchrakian N, Dhani NC, Jimenez W, Welch S, MacKay H, Ethier JL, Gilbert L, Li X, Rodriguez A, Chan L, Bowering V, Clarke B, Zhang T, King I, Downs G, Stockley T, Wang L, Udagani S, Oza AM, Lheureux S. Clinical outcome and biomarker assessments of a multi-centre phase II trial assessing niraparib with or without dostarlimab in recurrent endometrial carcinoma. Nat Commun 2023; 14:1452. [PMID: 36922497 PMCID: PMC10017680 DOI: 10.1038/s41467-023-37084-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
This multi-centre, non-randomized, open-label, phase II trial (NCT03016338), assessed niraparib monotherapy (cohort 1, C1), or niraparib and dostarlimab (cohort 2, C2) in patients with recurrent serous or endometrioid endometrial carcinoma. The primary endpoint was clinical benefit rate (CBR), with ≥5/22 overall considered of interest. Secondary outcomes were safety, objective response rate (ORR), duration of response, progression free survival and overall survival. Translational research was an exploratory outcome. Potential biomarkers were evaluated in archival tissue by immunohistochemistry and next generation sequencing panel. In C1, 25 patients were enrolled, and CBR was 20% (95% CI: 9-39) with median clinical benefit duration of 5.3 months. The ORR was 4% (95% CI: 0-20). In C2, 22 patients were enrolled, and the CBR was 31.8% (95% CI: 16-53) with median clinical benefit duration of 6.8 months. The ORR was 14% (95% CI: 3-35). No new safety signals were detected. No significant association was detected between clinical benefit and IHC markers (PTEN, p53, MMR, PD-L1), or molecular profiling (PTEN, TP53, homologous recombination repair genes). In conclusion, niraparib monotherapy did not meet the efficacy threshold. Niraparib in combination with dostarlimab showed modest activity.
Collapse
Affiliation(s)
- Ainhoa Madariaga
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
- Autonomous University of Barcelona, Barcelona, Spain
- Department of Medical Oncology, 12 de Octubre University Hospital, Madrid, Spain
| | - Swati Garg
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nairi Tchrakian
- University of Toronto, Toronto, ON, Canada
- Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada
| | - Neesha C Dhani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Waldo Jimenez
- Division of Gynecologic Oncology, Juravinski Cancer Centre, Hamilton, ON, Canada
| | - Stephen Welch
- Division of Medical Oncology and Hematology, London Health Sciences Center, London Regional Cancer Program, London, ON, Canada
| | - Helen MacKay
- Division of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Josee-Lyne Ethier
- Division of Medical Oncology and Hematology, Kingston Health Sciences Cancer Centre, Kingston, ON, Canada
| | - Lucy Gilbert
- Division of Gynecologic Oncology, McGill University Health Centre, Royal Victoria Hospital, Montréal, QC, Canada
| | - Xuan Li
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Angela Rodriguez
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Lucy Chan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Valerie Bowering
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Blaise Clarke
- University of Toronto, Toronto, ON, Canada
- Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada
| | - Tong Zhang
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Ian King
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gregory Downs
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tracy Stockley
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Lisa Wang
- University of Toronto, Toronto, ON, Canada
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Smitha Udagani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Stephanie Lheureux
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Madariaga A, Sanchez-Bayona R, Herrera FG, Ramirez PT, González Martín A. Outcomes and endpoints of relevance in gynecologic cancer clinical trials. Int J Gynecol Cancer 2023; 33:323-332. [PMID: 36878559 DOI: 10.1136/ijgc-2022-003727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Drug development is paramount to improve outcomes in patients with gynecologic cancers. A randomized clinical trial should measure whether a clinically relevant improvement is detected with the new intervention compared with the standard of care, using reproductible and appropriate endpoints. Clinically meaningful improvements in overall survival and/or quality of life (QoL) are the gold standards to measure benefit of new therapeutic strategies. Alternative endpoints, such as progression-free survival, provide an earlier measure of the effect of the new therapeutic drug, and are not confounded by the effect of subsequent lines of therapy. Yet, its surrogacy with improved overall survival or QoL is unclear in gynecologic malignancies. Of relevance to studies assessing maintenance strategies are other time-to-event endpoints, such as progression-free survival two and time to second subsequent treatment, which provide valuable information on the disease control in the longer term. Translational and biomarker studies are increasingly being incorporated into gynecologic oncology clinical trials, as they may allow understanding of the biology of the disease, resistance mechanisms, and enable a better selection of patients who might benefit from the new therapeutic strategy. Globally, the endpoint selection of a clinical trial will differ according to the type of study, population, disease setting, and type of therapeutic strategy. This review provides an overview of primary and secondary endpoint selection of relevance for gynecologic oncology clinical trials.
Collapse
Affiliation(s)
- Ainhoa Madariaga
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Fernanda G Herrera
- Department of Oncology, Radiation Oncology Service, Centre Hospitalier Universitaire Vaudois CHUV-UNIL, Ludwig Institute for Cancer Research and AGORA Cancer Research Center, Lausanne, Switzerland
| | - Pedro T Ramirez
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
28
|
Zhao Z, Xu H, Wei Y, Sun L, Song Y. Deubiquitylase PSMD14 inhibits autophagy to promote ovarian cancer progression via stabilization of LRPPRC. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166594. [PMID: 36328147 DOI: 10.1016/j.bbadis.2022.166594] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022]
Abstract
Autophagy is a key cellular process, which exists in many tumors and plays dual roles in tumor promotion and suppression. However, the role and mechanism of aberrant autophagy in ovarian cancer remains unclear. Ubiquitin-proteasome pathway is the most important pathway for specific protein degradation. Deubiquitinases (DUBs) have crucial roles in all the stages of tumorigenesis and progression. Herein, we explore the DUBs which contribute to aberrant autophagy in ovarian cancer. TCGA data analysis shows that the autophagy level is suppressed, and the selective autophagy receptor SQSTM1/p62 is abnormally high expressed in ovarian cancer. We screen and identify that the deubiquitinase PSMD14 negatively regulates autophagy level. Functional studies show that increased PSMD14 expression remarkably enhances ovarian cancer cells malignancy, whereas knockdown of PSMD14 has the opposite effect. Furthermore, in vivo assays show that knockdown of PSMD14 inhibits the growth, lung and abdominal metastasis of ovarian cancer. Mechanistically, PSMD14 directly interacts with LRPPRC and inhibits its ubiquitination, thereby inhibiting autophagy through LRPPRC/Beclin1-Bcl-2/SQSTM1 signaling pathway. Next, we demonstrate that PSMD14 is upregulated in ovarian cancer and high expression of PSMD14 positively correlates with LRPPRC. Taken together, we clarify the role of autophagy in regulating the ovarian cancer phenotype and provide insights into regulatory mechanism of autophagy in ovarian cancer.
Collapse
Affiliation(s)
- Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Heyang Xu
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuan Wei
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Sun
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
29
|
Kacsir I, Sipos A, Kiss T, Major E, Bajusz N, Tóth E, Buglyó P, Somsák L, Kardos G, Bai P, Bokor É. Half sandwich-type osmium, ruthenium, iridium and rhodium complexes with bidentate glycosyl heterocyclic ligands induce cytostasis in platinum-resistant ovarian cancer cells and bacteriostasis in Gram-positive multiresistant bacteria. Front Chem 2023; 11:1086267. [PMID: 36793764 PMCID: PMC9923724 DOI: 10.3389/fchem.2023.1086267] [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: 11/01/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
The toxicity of and resistance to platinum complexes as cisplatin, oxaliplatin or carboplatin calls for the replacement of these therapeutic agents in clinical settings. We have previously identified a set of half sandwich-type osmium, ruthenium and iridium complexes with bidentate glycosyl heterocyclic ligands exerting specific cytostatic activity on cancer cells but not on non-transformed primary cells. The apolar nature of the complexes, conferred by large, apolar benzoyl protective groups on the hydroxyl groups of the carbohydrate moiety, was the main molecular feature to induce cytostasis. We exchanged the benzoyl protective groups to straight chain alkanoyl groups with varying length (3 to 7 carbon units) that increased the IC50 value as compared to the benzoyl-protected complexes and rendered the complexes toxic. These results suggest a need for aromatic groups in the molecule. The pyridine moiety of the bidentate ligand was exchanged for a quinoline group to enlarge the apolar surface of the molecule. This modification decreased the IC50 value of the complexes. The complexes containing [(η6-p-cymene)Ru(II)], [(η6-p-cymene)Os(II)] or [(η5-Cp*)Ir(III)] were biologically active unlike the complex containing [(η5-Cp*)Rh(III)]. The complexes with cytostatic activity were active on ovarian cancer (A2780, ID8), pancreatic adenocarcinoma (Capan2), sarcoma (Saos) and lymphoma cell lines (L428), but not on primary dermal fibroblasts and their activity was dependent on reactive oxygen species production. Importantly, these complexes were cytostatic on cisplatin-resistant A2780 ovarian cancer cells with similar IC50 values as on cisplatin-sensitive A2780 cells. In addition, the quinoline-containing Ru and Os complexes and the short chain alkanoyl-modified complexes (C3 and C4) proved to be bacteriostatic in multiresistant Gram-positive Enterococcus and Staphylococcus aureus isolates. Hereby, we identified a set of complexes with submicromolar to low micromolar inhibitory constants against a wide range of cancer cells, including platinum resistant cells and against multiresistant Gram-positive bacteria.
Collapse
Affiliation(s)
- István Kacsir
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
- Doctoral School of Chemistry, University of Debrecen, Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Kiss
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Evelin Major
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Nikolett Bajusz
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Buglyó
- Department of Inorganic and Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, Hungary
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
30
|
LRRC superfamily expression in stromal cells predicts the clinical prognosis and platinum resistance of ovarian cancer. BMC Med Genomics 2023; 16:10. [PMID: 36653841 PMCID: PMC9850808 DOI: 10.1186/s12920-023-01435-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Leucine-rich repeat sequence domains are known to mediate protein‒protein interactions. Recently, some studies showed that members of the leucine rich repeat containing (LRRC) protein superfamily may become new targets for the diagnosis and treatment of tumours. However, it is not known whether any of the LRRC superfamily genes is expressed in the stroma of ovarian cancer (OC) and is associated with prognosis. METHODS The clinical data and transcriptional profiles of OC patients from the public databases TCGA (n = 427), GTEx (n = 88) and GEO (GSE40266 and GSE40595) were analysed by R software. A nomogram model was also generated through R. An online public database was used for auxiliary analysis of prognosis, immune infiltration and protein‒protein interaction (PPI) networks. Immunohistochemistry and qPCR were performed to determine the protein and mRNA levels of genes in high-grade serous ovarian cancer (HGSC) tissues of participants and the MRC-5 cell line induced by TGF-β. RESULTS LRRC15 and LRRC32 were identified as differentially expressed genes from the LRRC superfamily by GEO transcriptome analysis. PPI network analysis suggested that they were most enriched in TGF-β signalling. The TCGA-GTEx analysis results showed that only LRRC15 was highly expressed in both cancer-associated fibroblasts (CAFs) and the tumour stroma of OC and was related to clinical prognosis. Based on this, we developed a nomogram model to predict the incidence of adverse outcomes in OC. Moreover, LRRC15 was positively correlated with CAF infiltration and negatively correlated with CD8 + T-cell infiltration. As a single indicator, LRRC15 had the highest accuracy (AUC = 0.920) in predicting the outcome of primary platinum resistance. CONCLUSIONS The LRRC superfamily is related to the TGF-β pathway in the microenvironment of OC. LRRC15, as a stromal biomarker, can predict the clinical prognosis of HGSC and promote the immunosuppressive microenvironment. LRRC15 may be a potential therapeutic target for reversing primary resistance in OC.
Collapse
|
31
|
Multiparameter single-cell proteomic technologies give new insights into the biology of ovarian tumors. Semin Immunopathol 2023; 45:43-59. [PMID: 36635516 PMCID: PMC9974728 DOI: 10.1007/s00281-022-00979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy. Its diagnosis at advanced stage compounded with its excessive genomic and cellular heterogeneity make curative treatment challenging. Two critical therapeutic challenges to overcome are carboplatin resistance and lack of response to immunotherapy. Carboplatin resistance results from diverse cell autonomous mechanisms which operate in different combinations within and across tumors. The lack of response to immunotherapy is highly likely to be related to an immunosuppressive HGSOC tumor microenvironment which overrides any clinical benefit. Results from a number of studies, mainly using transcriptomics, indicate that the immune tumor microenvironment (iTME) plays a role in carboplatin response. However, in patients receiving treatment, the exact mechanistic details are unclear. During the past decade, multiplex single-cell proteomic technologies have come to the forefront of biomedical research. Mass cytometry or cytometry by time-of-flight, measures up to 60 parameters in single cells that are in suspension. Multiplex cellular imaging technologies allow simultaneous measurement of up to 60 proteins in single cells with spatial resolution and interrogation of cell-cell interactions. This review suggests that functional interplay between cell autonomous responses to carboplatin and the HGSOC immune tumor microenvironment could be clarified through the application of multiplex single-cell proteomic technologies. We conclude that for better clinical care, multiplex single-cell proteomic technologies could be an integral component of multimodal biomarker development that also includes genomics and radiomics. Collection of matched samples from patients before and on treatment will be critical to the success of these efforts.
Collapse
|
32
|
Pietkiewicz D, Plewa S, Zaborowski M, Garrett TJ, Matuszewska E, Kokot ZJ, Matysiak J. Mass spectrometry imaging in gynecological cancers: the best is yet to come. Cancer Cell Int 2022; 22:414. [PMID: 36536419 PMCID: PMC9764543 DOI: 10.1186/s12935-022-02832-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Mass spectrometry imaging (MSI) enables obtaining multidimensional results simultaneously in a single run, including regiospecificity and m/z values corresponding with specific proteins, peptides, lipids, etc. The knowledge obtained in this way allows for a multifaceted analysis of the studied issue, e.g., the specificity of the neoplastic process and the search for new therapeutic targets. Despite the enormous possibilities, this relatively new technique in many aspects still requires the development or standardization of analytical protocols (from collecting biological material, through sample preparation, analysis, and data collection, to data processing). The introduction of standardized protocols for MSI studies, with its current potential to extend diagnostic and prognostic capabilities, can revolutionize clinical pathology. As far as identifying ovarian cancer subtypes can be challenging, especially in poorly differentiated tumors, developing MSI-based algorithms may enhance determining prognosis and tumor staging without the need for extensive surgery and optimize the choice of subsequent therapy. MSI might bring new solutions in predicting response to treatment in patients with endometrial cancer. Therefore, MSI may help to revolutionize the future of gynecological oncology in terms of diagnostics, treatment, and predicting the response to therapy. This review will encompass several aspects, e.g., contemporary discoveries in gynecological cancer research utilizing MSI, indicates current challenges, and future perspectives on MSI.
Collapse
Affiliation(s)
- Dagmara Pietkiewicz
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznan, Poland.
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznan, Poland
| | - Mikołaj Zaborowski
- Gynecologic Oncology Department, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznan, Poland
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Eliza Matuszewska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznan, Poland
| | - Zenon J Kokot
- Faculty of Health Sciences, Calisia University, 13 Kaszubska Street, 62-800, Kalisz, Poland
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-806, Poznan, Poland
| |
Collapse
|
33
|
Tian L, Li X, Ji H, Yu Q, Yang M, Guo L, Huang L, Gao W. Melanin-like nanoparticles: advances in surface modification and tumour photothermal therapy. J Nanobiotechnology 2022; 20:485. [PMCID: PMC9675272 DOI: 10.1186/s12951-022-01698-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022] Open
Abstract
Currently, tumor treatments are characterized by intelligence, diversity and personalization, but the therapeutic reagents used are often limited in clinical efficacy due to problems with water solubility, targeting, stability and multidrug resistance. To remedy these shortcomings, the application of multifunctional nanotechnology in the biomedical field has been widely studied. Synthetic melanin nanoparticles (MNPs) surfaces which contain highly reactive chemical groups such as carboxyl, hydroxyl and amine groups, can be used as a reaction platform on which to graft different functional components. In addition, MNPs easily adhere to substrate surface, and serve as a secondary reaction platform to modify it. The multifunctionality and intrinsic biocompatibility make melanin-like nanoparticles promising as a multifunctional and powerful nanoplatform for oncological applications. This paper first reviews the preparation methods, polymerization mechanisms and physicochemical properties of melanin including natural melanin and chemically synthesized melanin to guide scholars in MNP-based design. Then, recent advances in MNPs especially synthetic polydopamine (PDA) melanin for various medical oncological applications are systematically and thoroughly described, mainly focusing on bioimaging, photothermal therapy (PTT), and drug delivery for tumor therapy. Finally, based on the investigated literature, the current challenges and future directions for clinical translation are reasonably discussed, focusing on the innovative design of MNPs and further elucidation of pharmacokinetics. This paper is a timely and comprehensive and detailed study of the progress of MNPs in tumor therapy, especially PTT, and provides ideas for the design of personalized and customizable oncology nanomedicines to address the heterogeneity of the tumor microenvironment.
Collapse
Affiliation(s)
- Luyao Tian
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Xia Li
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Haixia Ji
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Qing Yu
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Mingjuan Yang
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| | - Lanping Guo
- grid.410318.f0000 0004 0632 3409National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Luqi Huang
- grid.410318.f0000 0004 0632 3409National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Wenyuan Gao
- grid.33763.320000 0004 1761 2484Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300193 China
| |
Collapse
|
34
|
Madariaga A, Mitchell SA, Pittman T, Wang L, Bowering V, Kavak N, Quintos J, Chang K, Ramsahai J, Karakasis K, Welch SA, Dhani NC, Lheureux S, Oza AM. Patient self-reporting of tolerability using PRO-CTCAE in a randomized double-blind, placebo-controlled phase II trial comparing gemcitabine in combination with adavosertib or placebo in patients with platinum resistant or refractory epithelial ovarian carcinoma. Gynecol Oncol 2022; 167:226-233. [PMID: 36055813 PMCID: PMC10731422 DOI: 10.1016/j.ygyno.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND A double-blind, randomized, placebo-controlled, phase 2 trial assessed gemcitabine in combination with the wee1 inhibitor adavosertib or placebo in platinum resistant or refractory high grade serous ovarian cancer (HGSOC), demonstrating improved progression free and overall survival favouring the adavosertib/gemcitabine arm. An exploratory objective of the study included the PRO-CTCAE assessment, to capture self-reporting of frequency, severity and/or interference of symptomatic adverse events (syAEs). METHODS PRO-CTCAE items at baseline, days 1 and 15 of each cycle and off treatment, were completed in two centres, with the objective of characterizing syAEs in the first three months of therapy. The maximum post-baseline score proportion for each syAE was tabulated per patient. The 12-week area under the curve (AUC12w) as a measure of syAE over-time and incremental AUC12w (iAUC12w) for adjustment to baseline syAEs. RESULTS Sixty-one patients were approached for PRO-CTCAE surveys and 55 were evaluable. Among patients with HGSOC, 28 received gemcitabine/adavosertib (arm A) and 19 gemcitabine/placebo (arm B). Survey completion rates were high. The proportion of participants with positive (≥1) PRO-CTCAE scores was higher for difficulty swallowing with gemcitabine/adavosertib (arm A 35.7% vs arm B 5.3%, p = 0.02). The high score (≥3) syAEs showed more frequent diarrhea with gemcitabine/adavosertib (arm A 25% vs arm B 0%, p = 0.03). The proportions of worsening syAEs over time were higher in patients receiving gemcitabine/adavosertib for difficulty swallowing (arm A 35.7% vs arm B 5.3%; p = 0.03) and fatigue severity (arm A 71.43% vs arm B 42.1%; p = 0.04). CONCLUSIONS The longitudinal assessment of patient self-reported tolerability showed greater difficulty swallowing and fatigue severity in patients receiving gemcitabine/adavosertib, compared to gemcitabine/placebo. PRO-CTCAE provides complementary and objective assessment of drug tolerability from a patient's perspective.
Collapse
Affiliation(s)
- Ainhoa Madariaga
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada; Autonomous University of Barcelona, Barcelona, Spain
| | - Sandra A Mitchell
- National Cancer Institute at the National Institutes of Health, Rockville, USA
| | - Tyler Pittman
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lisa Wang
- University of Toronto, Toronto, Ontario, Canada; Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Valerie Bowering
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nisan Kavak
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Judy Quintos
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Karen Chang
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Janelle Ramsahai
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Katherine Karakasis
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Stephen A Welch
- Division of Medical Oncology & Hematology, London Health Science, London, Ontario, Canada
| | - Neesha C Dhani
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Lheureux
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Amit M Oza
- Division of Medical Oncology & Hematology, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Roering P, Siddiqui A, Heuser VD, Potdar S, Mikkonen P, Oikkonen J, Li Y, Pikkusaari S, Wennerberg K, Hynninen J, Grenman S, Huhtinen K, Auranen A, Carpén O, Kaipio K. Effects of Wee1 inhibitor adavosertib on patient-derived high-grade serous ovarian cancer cells are multiple and independent of homologous recombination status. Front Oncol 2022; 12:954430. [PMID: 36081565 PMCID: PMC9445195 DOI: 10.3389/fonc.2022.954430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Objective A major challenge in the treatment of platinum-resistant high-grade serous ovarian cancer (HGSOC) is lack of effective therapies. Much of ongoing research on drug candidates relies on HGSOC cell lines that are poorly documented. The goal of this study was to screen for effective, state-of-the-art drug candidates using primary HGSOC cells. In addition, our aim was to dissect the inhibitory activities of Wee1 inhibitor adavosertib on primary and conventional HGSOC cell lines. Methods A comprehensive drug sensitivity and resistance testing (DSRT) on 306 drug compounds was performed on three patient-derived genetically unique HGSOC cell lines and two commonly used ovarian cancer cell lines. The effect of adavosertib on the cell lines was tested in several assays, including cell-cycle analysis, apoptosis induction, proliferation, wound healing, DNA damage, and effect on nuclear integrity. Results Several compounds exerted cytotoxic activity toward all cell lines, when tested in both adherent and spheroid conditions. In further cytotoxicity tests, adavosertib exerted the most consistent cytotoxic activity. Adavosertib affected cell-cycle control in patient-derived and conventional HGSOC cells, inducing G2/M accumulation and reducing cyclin B1 levels. It induced apoptosis and inhibited proliferation and migration in all cell lines. Furthermore, the DNA damage marker γH2AX and the number of abnormal cell nuclei were clearly increased following adavosertib treatment. Based on the homologous recombination (HR) signature and functional HR assays of the cell lines, the effects of adavosertib were independent of the cells' HR status. Conclusion Our study indicates that Wee1 inhibitor adavosertib affects several critical functions related to proliferation, cell cycle and division, apoptosis, and invasion. Importantly, the effects are consistent in all tested cell lines, including primary HGSOC cells, and independent of the HR status of the cells. Wee1 inhibition may thus provide treatment opportunities especially for patients, whose cancer has acquired resistance to platinum-based chemotherapy or PARP inhibitors.
Collapse
Affiliation(s)
- Pia Roering
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
- *Correspondence: Pia Roering, ; Olli Carpén,
| | - Arafat Siddiqui
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Vanina D. Heuser
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Swapnil Potdar
- High Throughput Biomedicine Unit, Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Piia Mikkonen
- Helsinki Institute of Life Science (HiLIFE), Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jaana Oikkonen
- Research Program in Systems Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Yilin Li
- Research Program in Systems Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sanna Pikkusaari
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Krister Wennerberg
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Johanna Hynninen
- Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, Turku, Finland
| | - Seija Grenman
- Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, Turku, Finland
| | - Kaisa Huhtinen
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
- Research Program in Systems Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Annika Auranen
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Olli Carpén
- Department of Pathology, Precision Cancer Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- *Correspondence: Pia Roering, ; Olli Carpén,
| | - Katja Kaipio
- Institute of Biomedicine and Finnish Cancer Center (FICAN) West Cancer Centre, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
36
|
Awada A, Ahmad S, McKenzie ND, Holloway RW. Immunotherapy in the Treatment of Platinum-Resistant Ovarian Cancer: Current Perspectives. Onco Targets Ther 2022; 15:853-866. [PMID: 35982728 PMCID: PMC9379118 DOI: 10.2147/ott.s335936] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic cancer. The gold standard therapeutic approach is a combination of surgery plus chemotherapy. Unfortunately, 80% of patients with EOC suffer recurrence within 2-years and the overall response rate for platinum-resistant epithelial ovarian cancer to cytotoxic chemotherapy or poly-(adenosine diphosphate)-ribose polymerase (PARP) inhibitor is modest. New therapies are needed to improve overall survival. The role of immunotherapy has been established in endometrial and cervical cancers, however its effective use in EOC has been limited due to the intrinsic genomics and micro-immune environment associated with EOC. Studies evaluating immunotherapy, largely immune checkpoint inhibitors (ICI), have shown limited activity, yet some patients benefit greatly. Thus, significant efforts must be devoted to finding new strategies for the use of immunotherapy/immunomodulatory drugs (IMiDs). Immunotherapy has a well-tolerated safety profile; however, cost-effectiveness can be an obstacle. The aim of this article is to review the most recent research into the use of IMiDs in patients with platinum-resistant epithelial ovarian cancer.
Collapse
Affiliation(s)
- Ahmad Awada
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Nathalie D McKenzie
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Robert W Holloway
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| |
Collapse
|
37
|
Mahmood T, Ved A, Siddiqui MH, Ahsan F, Shamim A, Ansari VA, Ahmad A, Kashyap MK. An in-Depth Analysis of Ovarian Cancer: Pathogenesis and Clinical Manifestation. Drug Res (Stuttg) 2022; 72:424-434. [PMID: 35760337 DOI: 10.1055/a-1867-4654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Ovarian cancer is characterized by the establishment of tolerance, the recurrence of disease, as well as a poor prognosis. Gene signatures in ovarian cancer cells enable cancer medicine research, therapy, prevention, & management problematic. Notwithstanding advances in tumor puncture surgery, novel combinations regimens, and abdominal radiation, which can provide outstanding reaction times, the bulk of gynecological tumor patients suffer from side effects & relapse. As a consequence, more therapy alternatives for individuals with ovarian cancer must always be studied to minimize side effects and improve progression-free and total response rates. The development of cancer medications is presently undergoing a renaissance in the quest for descriptive and prognostic ovarian cancer biomarkers. Nevertheless, abnormalities in the BRCA2 or BRCA1 genes, a variety of hereditary predispositions, unexplained onset and progression, molecular tumor diversity, and illness staging can all compromise the responsiveness and accuracy of such indicators. As a result, current ovarian cancer treatments must be supplemented with broad-spectrum & customized targeted therapeutic approaches. The objective of this review is to highlight recent contributions to the knowledge of the interrelations between selected ovarian tumor markers, various perception signs, and biochemical and molecular signaling processes, as well as one's interpretation of much more targeted and effective treatment interventions.
Collapse
Affiliation(s)
- Tarique Mahmood
- Department of Pharmacy, Integral University, Dasauli, Lucknow, India
| | - Akash Ved
- Department of Pharmacy, Goel Institute of Pharmaceutical Sciences, Lucknow, India
| | | | - Farogh Ahsan
- Department of Pharmacy, Integral University, Dasauli, Lucknow, India
| | - Arshiya Shamim
- Department of Pharmacy, Integral University, Dasauli, Lucknow, India
| | | | - Afroz Ahmad
- Department of Pharmacy, Integral University, Dasauli, Lucknow, India
| | - Monu Kumar Kashyap
- Department of Pharmacy, Goel Institute of Pharmaceutical Sciences, Lucknow, India
| |
Collapse
|
38
|
Yuan H, Yu Q, Pang J, Chen Y, Sheng M, Tang W. The Value of the Stemness Index in Ovarian Cancer Prognosis. Genes (Basel) 2022; 13:genes13060993. [PMID: 35741755 PMCID: PMC9222264 DOI: 10.3390/genes13060993] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological malignancies. It is associated with a difficult diagnosis and poor prognosis. Our study aimed to analyze tumor stemness to determine the prognosis feature of patients with OC. At this job, we selected the gene expression and the clinical profiles of patients with OC in the TCGA database. We calculated the stemness index of each patient using the one-class logistic regression (OCLR) algorithm and performed correlation analysis with immune infiltration. We used consensus clustering methods to classify OC patients into different stemness subtypes and compared the differences in immune infiltration between them. Finally, we established a prognostic signature by Cox and LASSO regression analysis. We found a significant negative correlation between a high stemness index and immune score. Pathway analysis indicated that the differentially expressed genes (DEGs) from the low- and high-mRNAsi groups were enriched in multiple functions and pathways, such as protein digestion and absorption, the PI3K-Akt signaling pathway, and the TGF-β signaling pathway. By consensus cluster analysis, patients with OC were split into two stemness subtypes, with subtype II having a better prognosis and higher immune infiltration. Furthermore, we identified 11 key genes to construct the prognostic signature for patients with OC. Among these genes, the expression levels of nine, including SFRP2, MFAP4, CCDC80, COL16A1, DUSP1, VSTM2L, TGFBI, PXDN, and GAS1, were increased in the high-risk group. The analysis of the KM and ROC curves indicated that this prognostic signature had a great survival prediction ability and could independently predict the prognosis for patients with OC. We established a stemness index-related risk prognostic module for OC, which has prognostic-independent capabilities and is expected to improve the diagnosis and treatment of patients with OC.
Collapse
|
39
|
Can Schlafen 11 Help to Stratify Ovarian Cancer Patients Treated with DNA-Damaging Agents? Cancers (Basel) 2022; 14:cancers14102353. [PMID: 35625957 PMCID: PMC9139752 DOI: 10.3390/cancers14102353] [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: 03/25/2022] [Revised: 04/25/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Platinum-based chemotherapy has been the cornerstone of systemic treatment in ovarian cancer. Since no validated molecular predictive markers have been identified yet, the response to platinum-based chemotherapy has been evaluated clinically, based on platinum-free interval. The new promising marker Schlafen 11 seems to correlate with sensitivity or resistance to DNA-damaging agents, including platinum compounds or PARP inhibitors in various types of cancer. We provide background information about the function of Schlafen 11, its evaluation in tumor tissue, and its prevalence in ovarian cancer. We discuss the current evidence of the correlation of Schlafen 11 expression in ovarian cancer with treatment outcomes and the potential use of Schlafen 11 as the key predictive and prognostic marker that could help to better stratify ovarian cancer patients treated with platinum-based chemotherapy or PARP inhibitors. We also provide perspectives on future directions in the research on this promising marker.
Collapse
|
40
|
Marastoni S, Madariaga A, Pesic A, Nair SN, Li ZJ, Shalev Z, Ketela T, Colombo I, Mandilaras V, Cabanero M, Bruce JP, Li X, Garg S, Wang L, Chen EX, Gill S, Dhani NC, Zhang W, Pintilie M, Bowering V, Koritzinsky M, Rottapel R, Wouters BG, Oza AM, Joshua AM, Lheureux S. Repurposing Itraconazole and Hydroxychloroquine to Target Lysosomal Homeostasis in Epithelial Ovarian Cancer. CANCER RESEARCH COMMUNICATIONS 2022; 2:293-306. [PMID: 36875717 PMCID: PMC9981200 DOI: 10.1158/2767-9764.crc-22-0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/13/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022]
Abstract
Drug repurposing is an attractive option for oncology drug development. Itraconazole is an antifungal ergosterol synthesis inhibitor that has pleiotropic actions including cholesterol antagonism, inhibition of Hedgehog and mTOR pathways. We tested a panel of 28 epithelial ovarian cancer (EOC) cell lines with itraconazole to define its spectrum of activity. To identify synthetic lethality in combination with itraconazole, a whole-genome drop-out genome-scale clustered regularly interspaced short palindromic repeats sensitivity screen in two cell lines (TOV1946 and OVCAR5) was performed. On this basis, we conducted a phase I dose-escalation study assessing the combination of itraconazole and hydroxychloroquine in patients with platinum refractory EOC (NCT03081702). We identified a wide spectrum of sensitivity to itraconazole across the EOC cell lines. Pathway analysis showed significant involvement of lysosomal compartments, the trans-golgi network and late endosomes/lysosomes; similar pathways are phenocopied by the autophagy inhibitor, chloroquine. We then demonstrated that the combination of itraconazole and chloroquine displayed Bliss defined synergy in EOC cancer cell lines. Furthermore, there was an association of cytotoxic synergy with the ability to induce functional lysosome dysfunction, by chloroquine. Within the clinical trial, 11 patients received at least one cycle of itraconazole and hydroxychloroquine. Treatment was safe and feasible with the recommended phase II dose of 300 and 600 mg twice daily, respectively. No objective responses were detected. Pharmacodynamic measurements on serial biopsies demonstrated limited pharmacodynamic impact. In vitro, itraconazole and chloroquine have synergistic activity and exert a potent antitumor effect by affecting lysosomal function. The drug combination had no clinical antitumor activity in dose escalation. Significance The combination of the antifungal drug itraconazole with antimalarial drug hydroxychloroquine leads to a cytotoxic lysosomal dysfunction, supporting the rational for further research on lysosomal targeting in ovarian cancer.
Collapse
Affiliation(s)
- Stefano Marastoni
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ainhoa Madariaga
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Autonomous University of Barcelona, Barcelona, Spain
| | - Aleksandra Pesic
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sree Narayanan Nair
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Zhu Juan Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Zvi Shalev
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ilaria Colombo
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Victoria Mandilaras
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Michael Cabanero
- Department of Pathology, Toronto General Hospital, Toronto, Ontario, Canada
| | - Jeff P Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Xuan Li
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Swati Garg
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Lisa Wang
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Eric X Chen
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sarbjot Gill
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Neesha C Dhani
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Wenjiang Zhang
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Valerie Bowering
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Bradly G Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Amit M Oza
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Anthony M Joshua
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Kinghorn Cancer Centre, Department of Medical Oncology, St Vincents Hospital, Sydney, Australia.,Garvan Institute of Medical Research, Sydney, Australia
| | - Stephanie Lheureux
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Qin C, Wu M, Wang X, Zhang W, Qi G, Wu NY, Liu X, Lu Y, Zhang J, Chai Y. Study on the mechanism of Danshen-Guizhi drug pair in the treatment of ovarian cancer based on network pharmacology and in vitro experiment. PeerJ 2022; 10:e13148. [PMID: 35411258 PMCID: PMC8994495 DOI: 10.7717/peerj.13148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/01/2022] [Indexed: 01/12/2023] Open
Abstract
Our study aims to explore the active components and mechanisms of the Danshen-Guizhi drug pair in treating ovarian cancer by network pharmacology and in vitro experiment. The "component-target-pathway" diagram of the Danshen-Guizhi drug pair was established by network pharmacology, and the effective active components, important targets as well as potential mechanisms of the Danshen-Guizhi drug pair were analyzed. The predicted results were verified by molecular docking and in vitro experiments. The main active components of the Danshen-Guizhi drug pair in the treatment of ovarian cancer are salviolone, luteolin, β-sitosterol and tanshinone IIA. The main core target is PTGS2. The pathways involved mainly include the cancer pathway, PI3K-Akt signaling pathway, and IL-17 signaling pathway. The molecular docking results showed that salviolone and tanshinone IIA had good binding ability to the target. The expression of PTGS2 mRNA and PGE2 in ovarian cells were significantly inhibited by salviolone. The mechanism of the Danshen-Guizhi drug pair in the treatment of ovarian cancer may be regulating cell proliferation, apoptosis and tumor immunity. This provides a theoretical basis for the clinical development and application of the Danshen-Guizhi drug pair.
Collapse
Affiliation(s)
- Chongzhen Qin
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Menglin Wu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinru Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenda Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangzhao Qi
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na-Yi Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaoting Liu
- The Second People’s Hospital of Hunan Province, Changsha, China
| | - Yaoyao Lu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingmin Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuna Chai
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Oh M, Son C, Rho SB, Kim M, Park K, Song SY. Stem Cell Factor SOX9 Interacts with a Cell Death Regulator RIPK1 and Results in Escape of Cancer Stem Cell Death. Cells 2022; 11:cells11030363. [PMID: 35159173 PMCID: PMC8834197 DOI: 10.3390/cells11030363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade ovarian cancer (HGOC) is the most lethal gynecological cancer, with high metastasis and recurrence. Cancer stem cells (CSCs) are responsible for its apoptosis resistance, cancer metastasis, and recurrence. Thus, targeting CSCs would be a promising strategy for overcoming chemotherapy resistance and improving patient prognosis in HGOC. Among upregulated oncogenic proteins in HGOC, we found that transcription factor SOX9 showed a strong correlation with stemness-regulating ALDH1A1 and was localized predominantly in the cytoplasm of HGOC with lymph node metastasis. In order to address the role of unusual cytoplasmic SOX9 and to explore its underlying mechanism in HGOC malignancy, a Y2H assay was used to identify a necroptotic cell death-associated cytoplasmic protein, receptor-interacting serine/threonine protein kinase 1 (RIPK1), as a novel SOX9-interacting partner and further mapped their respective interacting domains. The C-terminal region containing the transactivation domain of SOX9 interacted with the death domain of R1PK1. Consistent with its stemness-promoting function, SOX9 knockdown in vitro resulted in changes in cell morphology, cell cycle, stem cell marker expression, cell invasion, and sphere formation. Furthermore, in vivo knockdown completely inhibited tumor growth in mouse xenograft model. We propose that cytoplasmic SOX9-mediated cell death suppression would contribute to cancer stem cell survival in HGOC.
Collapse
Affiliation(s)
- Mijung Oh
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Chaeyeon Son
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
| | - Seung Bae Rho
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea;
| | - Minjeong Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Kyoungsook Park
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
- Correspondence: (K.P.); (S.Y.S.); Tel.: +82-10-8718-3625 (K.P.); +82-10-9933-2803 (S.Y.S.)
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
- Correspondence: (K.P.); (S.Y.S.); Tel.: +82-10-8718-3625 (K.P.); +82-10-9933-2803 (S.Y.S.)
| |
Collapse
|
43
|
Hyeraci M, Agnarelli L, Labella L, Marchetti F, Di Paolo ML, Samaritani S, Dalla Via L. trans-Dichloro(triphenylarsino)(N,N-dialkylamino)platinum(II) Complexes: In Search of New Scaffolds to Circumvent Cisplatin Resistance. Molecules 2022; 27:molecules27030644. [PMID: 35163916 PMCID: PMC8838190 DOI: 10.3390/molecules27030644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/01/2023] Open
Abstract
The high incidence of the resistance phenomenon represents one of the most important limitations to the clinical usefulness of cisplatin as an anticancer drug. Notwithstanding the considerable efforts to solve this problem, the circumvention of cisplatin resistance remains a challenge in the treatment of cancer. In this work, the synthesis and characterization of two trans-dichloro(triphenylarsino)(N,N-dialkylamino)platinum(II) complexes (1 and 2) were described. The trypan blue exclusion assay demonstrated an interesting antiproliferative effect for complex 1 in ovarian carcinoma-resistant cells, A2780cis. Quantitative analysis performed by ICP-AES demonstrated a scarce ability to platinate DNA, and a significant intracellular accumulation. The investigation of the mechanism of action highlighted the ability of 1 to inhibit the relaxation of supercoiled plasmid DNA mediated by topoisomerase II and to stabilize the cleavable complex. Cytofluorimetric analyses indicated the activation of the apoptotic pathway and the mitochondrial membrane depolarization. Therefore, topoisomerase II and mitochondria could represent possible intracellular targets. The biological properties of 1 and 2 were compared to those of the related trans-dichloro(triphenylphosphino)(N,N-dialkylamino)platinum(II) complexes in order to draw structure–activity relationships useful to face the resistance phenotype.
Collapse
Affiliation(s)
- Mariafrancesca Hyeraci
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Laura Agnarelli
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy; (L.A.); (L.L.); (F.M.); (S.S.)
| | - Luca Labella
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy; (L.A.); (L.L.); (F.M.); (S.S.)
- CISUP—Center for the Integration of Scientific Instruments, University of Pisa, 56126 Pisa, Italy
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy; (L.A.); (L.L.); (F.M.); (S.S.)
| | - Maria Luisa Di Paolo
- Department of Molecular Medicine, Università degli Studi di Padova, Via G. Colombo 3, 35131 Padova, Italy;
| | - Simona Samaritani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy; (L.A.); (L.L.); (F.M.); (S.S.)
- CISUP—Center for the Integration of Scientific Instruments, University of Pisa, 56126 Pisa, Italy
| | - Lisa Dalla Via
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Via F. Marzolo 5, 35131 Padova, Italy;
- Correspondence: ; Tel.: +39-049-8275712
| |
Collapse
|
44
|
Reactive Oxygen Species Production Is Responsible for Antineoplastic Activity of Osmium, Ruthenium, Iridium and Rhodium Half-Sandwich Type Complexes with Bidentate Glycosyl Heterocyclic Ligands in Various Cancer Cell Models. Int J Mol Sci 2022; 23:ijms23020813. [PMID: 35054999 PMCID: PMC8776094 DOI: 10.3390/ijms23020813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/04/2023] Open
Abstract
Platinum complexes are used in chemotherapy, primarily as antineoplastic agents. In this study, we assessed the cytotoxic and cytostatic properties of a set of osmium(II), ruthenium(II), iridium(III) and rhodium(III) half-sandwich-type complexes with bidentate monosaccharide ligands. We identified 5 compounds with moderate to negligible acute cytotoxicity but with potent long-term cytostatic activity. These structure-activity relationship studies revealed that: (1) osmium(II) p-cymene complexes were active in all models, while rhodium(III) and iridium(III) Cp* complexes proved largely inactive; (2) the biological effect was influenced by the nature of the central azole ring of the ligands—1,2,3-triazole was the most effective, followed by 1,3,4-oxadiazole, while the isomeric 1,2,4-oxadiazole abolished the cytostatic activity; (3) we found a correlation between the hydrophobic character of the complexes and their cytostatic activity: compounds with O-benzoyl protective groups on the carbohydrate moiety were active, compared to O-deprotected ones. The best compound, an osmium(II) complex, had an IC50 value of 0.70 µM. Furthermore, the steepness of the inhibitory curve of the active complexes suggested cooperative binding; cooperative molecules were better inhibitors than non-cooperative ones. The cytostatic activity of the active complexes was abolished by a lipid-soluble antioxidant, vitamin E, suggesting that oxidative stress plays a major role in the biological activity of the complexes. The complexes were active on ovarian cancer, pancreatic adenocarcinoma, osteosarcoma and Hodgkin’s lymphoma cells, but were inactive on primary, non-transformed human fibroblasts, indicating their applicability as potential anticancer agents.
Collapse
|
45
|
Azzalini E, Tierno D, Bartoletti M, Barbazza R, Giorda G, Puglisi F, Cecere SC, Losito NS, Russo D, Stanta G, Canzonieri V, Bonin S. AKT Isoforms Interplay in High-Grade Serous Ovarian Cancer Prognosis and Characterization. Cancers (Basel) 2022; 14:cancers14020304. [PMID: 35053468 PMCID: PMC8773580 DOI: 10.3390/cancers14020304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary New therapeutical strategies are needed to improve survival in high-grade serous ovarian cancer (HGSOC) patients. AKT inhibitors are promising agents able to act in synergy with PARP inhibitors and platinum-based therapies, but the subset of patients who could benefit from this approach is still unclear. We analyzed AKT isoforms expression in a retrospective cohort and we identified four AKT expression groups related to patients’ survival, tumor morphology and the BRCA status that could help in stratifying patients for future clinical trials. Abstract High-grade serous ovarian cancer (HGSOC) is among the deadliest gynecological malignancies. The acquired resistance to platinum-based therapies and the intrinsic heterogeneity of the disease contribute to the low survival rate. To improve patients’ outcomes, new combinatorial approaches able to target different tumor vulnerabilities and enhance the efficacy of the current therapies are required. AKT inhibitors are promising antineoplastic agents able to act in synergy with PARP inhibitors, but the spectrum of patients who can benefit from this combination is unclear, since the role of the three different isoforms of AKT is still unknown. Here, we study the expression of AKT isoforms on a retrospective cohort of archive tissue by RT-droplet digital PCR (ddPCR) analyzing their association with the clinicopathological features of patients. Based on AKT1/AKT2 and AKT1/AKT3 ratios, we define four AKT classes which were related to patients’ survival, tumor morphology and BRCA1 expression. Moreover, our results show that high AKT3 expression levels were frequently associated with tumors having classic features, a low number of mitoses and the presence of psammoma bodies. Overall, our study obtains new insights on AKT isoforms and their associations with the clinicopathological features of HGSOC patients. These evidences could help to better define the subsets of patients who can benefit from AKT and PARP inhibitors therapy in future clinical trials.
Collapse
Affiliation(s)
- Eros Azzalini
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, 33081 Aviano, Italy
| | - Domenico Tierno
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
| | - Michele Bartoletti
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, IRCCS CRO Aviano-National Cancer Institute, 33081 Aviano, Italy; (M.B.); (F.P.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Renzo Barbazza
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
| | - Giorgio Giorda
- Unit of Gynecologic Oncology Surgery, IRCCS CRO Aviano, National Cancer Institute, 33081 Aviano, Italy;
| | - Fabio Puglisi
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, IRCCS CRO Aviano-National Cancer Institute, 33081 Aviano, Italy; (M.B.); (F.P.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Sabrina Chiara Cecere
- Istituto Nazionale Tumori IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.C.C.); (N.S.L.); (D.R.)
| | - Nunzia Simona Losito
- Istituto Nazionale Tumori IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.C.C.); (N.S.L.); (D.R.)
| | - Daniela Russo
- Istituto Nazionale Tumori IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.C.C.); (N.S.L.); (D.R.)
| | - Giorgio Stanta
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
| | - Vincenzo Canzonieri
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, 33081 Aviano, Italy
- Correspondence: (V.C.); (S.B.); Tel.: +39−0434−659−618 (V.C.); +39−040−399−3266 (S.B.)
| | - Serena Bonin
- Department of Medical Sciences (DSM), University of Trieste, 34147 Trieste, Italy; (E.A.); (D.T.); (R.B.); (G.S.)
- Correspondence: (V.C.); (S.B.); Tel.: +39−0434−659−618 (V.C.); +39−040−399−3266 (S.B.)
| |
Collapse
|
46
|
El Bairi K, Singh S, Le Page C. Revisiting platinum-resistant ovarian cancer: Advances in therapy, molecular biomarkers, and clinical outcomes. Semin Cancer Biol 2021; 77:1-2. [PMID: 34492299 DOI: 10.1016/j.semcancer.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, United States; Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, United States; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, 36688, United States
| | - Cécile Le Page
- Research Institute of McGill University Health Center (RI-MUHC), Montréal, QC, Canada
| |
Collapse
|
47
|
Madariaga A, Bhat G, Wilson MK, Li X, Cyriac S, Bowering V, Hunt W, Gutierrez D, Bonilla L, Kasherman L, McMullen M, Wang L, Ghai S, Dhani NC, Oza AM, Lheureux S. Research biopsies in patients with gynecologic cancers: patient-reported outcomes, perceptions, and preferences. Am J Obstet Gynecol 2021; 225:658.e1-658.e9. [PMID: 34174204 DOI: 10.1016/j.ajog.2021.06.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite the growing integration of mandatory biopsies for correlative endpoints within oncology clinical trials, there are sparse data on patient-reported outcomes, perceptions, and preferences. OBJECTIVE This study aimed to prospectively assess the impact of research biopsies on the quality of life in patients with gynecologic cancer, evaluate patient-reported outcomes, and determine factors associated with patients' willingness to undergo sequential biopsies. STUDY DESIGN We conducted a prospective study in patients with gynecologic malignancies undergoing research biopsies between 2015 and 2019 at Princess Margaret Cancer Centre (ClinicalTrials.gov Identifier: NCT02334761). Here, we report the results of the paper-based surveys performed before and 1 week after biopsy. Although the questionnaires each assessed the impact of anxiety using a modified version of the Hospital Anxiety and Depression Scale, the postbiopsy questionnaire specifically assessed the likelihood of future biopsies, postbiopsy symptoms, complications, and perceptions. RESULTS A total of 129 patients were enrolled, of which 91 (70.5%) completed at least 1 questionnaire. These patients had either ovarian (89%; 81 of 91) or endometrial cancer (11%; 10 of 91). Of all biopsies taken, 75% were from the abdomen or pelvis (67 of 89). There was 1 clinician-reported complication, a perihepatic hematoma (1%). Pain during the biopsy and physical discomfort were experienced by 60.3% (41 of 68) and 61.8% (42 of 68), respectively. Embarrassment and loss of dignity were experienced by 13.2% (9 of 68) and 11.8% (8 of 68), respectively. Although the mean Hospital Anxiety and Depression Scale score was in the normal range before and after biopsy, there was a significant decline in the total score after the biopsy (prebiopsy, 5.3 [standard deviation, 4.7] vs postbiopsy, 3.7 [standard deviation, 4.5]; P=.005); 84% of subjects (58 of 69) stated that they would definitely or likely consent to another biopsy. There was no impact on patients' willingness for future biopsies based on Eastern Cooperative Oncology Group status, biopsy site, age, number of cores, and pain during the biopsy; however, subjects who reported feeling physically uncomfortable (odds ratio, 0.14; P=.005), embarrassed (odds ratio, 0.03; P=.004) or experienced loss of dignity (odds ratio, 0.05; P=.01) during the biopsy and those who experienced flu-like symptoms (odds ratio, 0.2; P=.018) or felt feverish (odds ratio, 0.2; P=.035) 1 week after biopsy, were less likely to undergo a sequential biopsy. Similarly, those with higher Hospital Anxiety and Depression Scale scores before biopsy (odds ratio, 0.83; P=.008) and after biopsy (odds ratio, 0.8; P=.003) were less likely to consent for another biopsy. CONCLUSION Research biopsies were generally well accepted. Most patients (83%) were willing to undergo serial biopsies if necessary. Addressing the potentially modifiable psychosocial aspects of the procedure may improve the experience with research biopsies for patients with gynecologic cancers.
Collapse
Affiliation(s)
- Ainhoa Madariaga
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada; Autonomous University of Barcelona, Barcelona, Spain
| | - Gita Bhat
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Michelle K Wilson
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Xuan Li
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sunu Cyriac
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Valerie Bowering
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Wendy Hunt
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - David Gutierrez
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Luisa Bonilla
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Lawrence Kasherman
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Michelle McMullen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Lisa Wang
- University of Toronto, Toronto, Ontario, Canada; Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sangeet Ghai
- University of Toronto, Toronto, Ontario, Canada; Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Neesha C Dhani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Lheureux
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
48
|
Ghoneum A, Almousa S, Warren B, Abdulfattah AY, Shu J, Abouelfadl H, Gonzalez D, Livingston C, Said N. Exploring the clinical value of tumor microenvironment in platinum-resistant ovarian cancer. Semin Cancer Biol 2021; 77:83-98. [PMID: 33476723 PMCID: PMC8286277 DOI: 10.1016/j.semcancer.2020.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Platinum resistance in epithelial ovarian cancer (OvCa) is rising at an alarming rate, with recurrence of chemo-resistant high grade serous OvCa (HGSC) in roughly 75 % of all patients. Additionally, HGSC has an abysmal five-year survival rate, standing at 39 % and 17 % for FIGO stages III and IV, respectively. Herein we review the crucial cellular interactions between HGSC cells and the cellular and non-cellular components of the unique peritoneal tumor microenvironment (TME). We highlight the role of the extracellular matrix (ECM), ascitic fluid as well as the mesothelial cells, tumor associated macrophages, neutrophils, adipocytes and fibroblasts in platinum-resistance. Moreover, we underscore the importance of other immune-cell players in conferring resistance, including natural killer cells, myeloid-derived suppressive cells (MDSCs) and T-regulatory cells. We show the clinical relevance of the key platinum-resistant markers and their correlation with the major pathways perturbed in OvCa. In parallel, we discuss the effect of immunotherapies in re-sensitizing platinum-resistant patients to platinum-based drugs. Through detailed analysis of platinum-resistance in HGSC, we hope to advance the development of more effective therapy options for this aggressive disease.
Collapse
Affiliation(s)
- Alia Ghoneum
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Sameh Almousa
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Bailey Warren
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Ammar Yasser Abdulfattah
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Alexandria University School of Medicine, Alexandria, Egypt
| | - Junjun Shu
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; The Third Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hebatullah Abouelfadl
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Department of Genetics, Animal Health Research Institute, Dokki, Egypt
| | - Daniela Gonzalez
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Christopher Livingston
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Neveen Said
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Comprehensive Cancer Center, Winston Salem, NC, 27157, USA.
| |
Collapse
|
49
|
Kacsir I, Sipos A, Ujlaki G, Buglyó P, Somsák L, Bai P, Bokor É. Ruthenium Half-Sandwich Type Complexes with Bidentate Monosaccharide Ligands Show Antineoplastic Activity in Ovarian Cancer Cell Models through Reactive Oxygen Species Production. Int J Mol Sci 2021; 22:ijms221910454. [PMID: 34638791 PMCID: PMC8508960 DOI: 10.3390/ijms221910454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022] Open
Abstract
Ruthenium complexes are developed as substitutes for platinum complexes to be used in the chemotherapy of hematological and gynecological malignancies, such as ovarian cancer. We synthesized and screened 14 ruthenium half-sandwich complexes with bidentate monosaccharide ligands in ovarian cancer cell models. Four complexes were cytostatic, but not cytotoxic on A2780 and ID8 cells. The IC50 values were in the low micromolar range (the best being 0.87 µM) and were similar to or lower than those of the clinically available platinum complexes. The active complexes were cytostatic in cell models of glioblastoma, breast cancer, and pancreatic adenocarcinoma, while they were not cytostatic on non-transformed human skin fibroblasts. The bioactive ruthenium complexes showed cooperative binding to yet unidentified cellular target(s), and their activity was dependent on reactive oxygen species production. Large hydrophobic protective groups on the hydroxyl groups of the sugar moiety were needed for biological activity. The cytostatic activity of the ruthenium complexes was dependent on reactive species production. Rucaparib, a PARP inhibitor, potentiated the effects of ruthenium complexes.
Collapse
Affiliation(s)
- István Kacsir
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary; (I.K.); (L.S.)
- Doctoral School of Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.S.); (G.U.)
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.S.); (G.U.)
| | - Péter Buglyó
- Department of Inorganic & Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary; (I.K.); (L.S.)
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.S.); (G.U.)
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, H-4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: (P.B.); (É.B.)
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary; (I.K.); (L.S.)
- Correspondence: (P.B.); (É.B.)
| |
Collapse
|
50
|
Serini S, Cassano R, Bruni M, Servidio C, Calviello G, Trombino S. Characterization of a hyaluronic acid and folic acid-based hydrogel for cisplatin delivery: Antineoplastic effect in human ovarian cancer cells in vitro. Int J Pharm 2021; 606:120899. [PMID: 34324990 DOI: 10.1016/j.ijpharm.2021.120899] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 01/01/2023]
Abstract
We successfully prepared and characterized a hyaluronic acid- and folic acid-based hydrogel for the delivery of cisplatin (GEL-CIS) with the aim to induce specific and efficient incorporation of CIS into ovarian cancer (OC) cells, improve its antineoplastic effect and avoid CIS-resistance. The slow and controlled release of the drug from the polymeric network and its swelling degree at physiologic pH suggested its suitability for CIS delivery in OC. We compared here the effects of pure CIS to that of GEL-CIS on human OC cell lines, either wild type or CIS-resistant, in basal conditions and in the presence of macrophage-derived conditioned medium, mimicking the action of tumor-associated macrophages in vivo. GEL-CIS inhibited OC cell growth and migration more efficiently than pure CIS and modulated the expression of proteins involved in the Epithelial Mesenchymal Transition, a process playing a key role in OC metastatic spread and resistance to CIS.
Collapse
Affiliation(s)
- Simona Serini
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy
| | - Roberta Cassano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Matilde Bruni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Camilla Servidio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Gabriella Calviello
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy.
| | - Sonia Trombino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| |
Collapse
|