1
|
Saha N, Lee SG, Brockmann EC, de la Cruz MJ, Goldgur Y, Mendoza RP, Stanchina ED, Love TM, Marvald J, Xu Y, Xu K, Himanen JP, Lamminmäki U, Veach D, Nikolov DB. Fully human monoclonal antibody targeting the cysteine-rich substrate-interacting region of ADAM17 on cancer cells. Biomed Pharmacother 2024; 180:117605. [PMID: 39461016 DOI: 10.1016/j.biopha.2024.117605] [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: 08/08/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
ADAM17 sheds EGFR/erbB ligands and triggers oncogenic pathways that lead to the progression of solid tumors. We targeted the ADAM17 disintegrin and cysteine rich domain region (D+C) to generate a panel of single-chain antibody fragments (scFvs) that selectively bind to the D or C domains of ADAM17, but not of ADAM10 or ADAM19. From the panel, we selected one scFv, referred to as C12, based on its high binding affinity towards the target, and re-formatted it to a full IgG for further studies. High-resolution cryo-electron microscopy studies documented that the mAb binds to the ADAM17 C-domain that in ADAM proteases, notably ADAM10 and ADAM17, is known to impart substrate-specificity. The C12 mAb significantly inhibited EGFR phosphorylation in cancer cell lines by hindering the cleavage of EGFR ligands tethered to the cell surface. This inhibition provides a mechanism for potential anti-tumor effects, and indeed C12 diminished the viability of a variety of EGFR-expressing cancer cell lines. Cell-based ELISA studies revealed that C12 preferentially bound to activated ADAM17 present on tumor cells, as compared to the autoinhibited ADAM17 that is the predominant form on HEK293 and other non-tumor cells. C12 also exhibited tumor growth inhibition in an ovarian cancer xenograft mouse model. Consistent with its selective tumor cell binding in vitro, radioimmuno PET (positron emission tomography) imaging with 89Zr-DFO-C12 in mouse xenograft models confirmed tumoral accumulation of the C12 mAb.
Collapse
Affiliation(s)
- Nayanendu Saha
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Sang Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | | | - M Jason de la Cruz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Rachelle P Mendoza
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Elisa de Stanchina
- Antitumor Assessment Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Tanzy M Love
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Josh Marvald
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - Yan Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Kai Xu
- Texas Therapeutic Institute, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Juha P Himanen
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Urpo Lamminmäki
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Dimitar B Nikolov
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
2
|
Wang ZB, Zhang X, Fang C, Liu XT, Liao QJ, Wu N, Wang J. Immunotherapy and the ovarian cancer microenvironment: Exploring potential strategies for enhanced treatment efficacy. Immunology 2024; 173:14-32. [PMID: 38618976 DOI: 10.1111/imm.13793] [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: 09/15/2023] [Accepted: 04/05/2024] [Indexed: 04/16/2024] Open
Abstract
Despite progress in cancer immunotherapy, ovarian cancer (OC) prognosis continues to be disappointing. Recent studies have shed light on how not just tumour cells, but also the complex tumour microenvironment, contribute to this unfavourable outcome of OC immunotherapy. The complexities of the immune microenvironment categorize OC as a 'cold tumour'. Nonetheless, understanding the precise mechanisms through which the microenvironment influences the effectiveness of OC immunotherapy remains an ongoing scientific endeavour. This review primarily aims to dissect the inherent characteristics and behaviours of diverse cells within the immune microenvironment, along with an exploration into its reprogramming and metabolic changes. It is expected that these insights will elucidate the operational dynamics of the immune microenvironment in OC and lay a theoretical groundwork for improving the efficacy of immunotherapy in OC management.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Xiu Zhang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Chao Fang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Xiao-Ting Liu
- The Second People's Hospital of Hunan Province, Changsha, China
| | - Qian-Jin Liao
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Nayiyuan Wu
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Jing Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| |
Collapse
|
3
|
Takasaki K, Ichinose T, Nishida H, Miyagawa Y, Hashimoto K, Watanabe S, Takahashi Y, Hirano M, Hiraike H, Sasajima Y, Nagasaka K. Prognostic analysis of peritoneal washing cytology during interval debulking surgery in advanced ovarian cancer. J Ovarian Res 2024; 17:170. [PMID: 39182152 PMCID: PMC11344424 DOI: 10.1186/s13048-024-01494-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Interval debulking surgery (IDS) following neoadjuvant chemotherapy is a treatment option for advanced ovarian cancer. Optimal surgery is required for better survival; however, while peritoneal washing cytology (PWC) has been identified as a prognostic factor, its comprehensive assessment during IDS remains unexplored. Therefore, we aimed to evaluate PWC efficacy during IDS, alongside other factors including residual disease and the modeled cancer antigen 125 (CA-125) ELIMination rate constant K (KELIM), by retrospectively reviewing the medical records of 25 patients with advanced ovarian cancer underwent neoadjuvant chemotherapy and IDS between January 2017 to June 2023. RESULTS Twelve (48.0%) patients were PWC-positive, and the remainder were PWC-negative. PWC was performed at laparotomy during IDS, after which five (41.7%) PWC-positive and four (30.8%) PWC-negative patients received bevacizumab, an anti-vascular endothelial growth factor monoclonal antibody, for maintenance treatment. Four (33.3%) PWC-positive and 10 (76.9%) PWC-negative patients received poly adenosine diphosphate (ADP)-ribose polymerase inhibitors. In patients who received bevacizumab and poly ADP-ribose polymerase inhibitors, overall survival and progression-free survival did not significantly differ between those who were PWC-positive and PWC-negative (p = 0.27 and 0.20, respectively). Progression-free survival significantly differed between those with favorable and unfavorable CA-125 KELIM (p = 0.02). Multivariate analysis indicated that optimal surgery and favorable CA-125 KELIM were associated with better progression-free survival (p < 0.01 and 0.02, respectively), with only optimal surgery associated with better overall survival (p = 0.04). CONCLUSIONS A positive PWC at IDS was not associated with survival in advanced ovarian cancer. Our findings indicate that although PWC status at IDS should be one of the factors determining survival in patients with advanced ovarian cancer, recent improvements in maintenance therapy may make the combination of CA-125 KELIM and PWC status a more useful prognostic factor in selecting treatment after IDS. Further studies are needed to validate these results, highlighting the potential importance of maintenance treatment after IDS and the need for further research to validate the clinical significance of a positive PWC.
Collapse
Affiliation(s)
- Kazuki Takasaki
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Takayuki Ichinose
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Haruka Nishida
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yuko Miyagawa
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Kei Hashimoto
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Saya Watanabe
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yuko Takahashi
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Mana Hirano
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Haruko Hiraike
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yuko Sasajima
- Department of Pathology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Kazunori Nagasaka
- Department of Obstetrics and Gynecology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605, Japan.
| |
Collapse
|
4
|
Tocci P, Roman C, Sestito R, Caprara V, Sacconi A, Molineris I, Tonon G, Blandino G, Bagnato A. The endothelin-1-driven tumor-stroma feed-forward loops in high-grade serous ovarian cancer. Clin Sci (Lond) 2024; 138:851-862. [PMID: 38884602 PMCID: PMC11230866 DOI: 10.1042/cs20240346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/18/2024]
Abstract
The high-grade serous ovarian cancer (HG-SOC) tumor microenvironment (TME) is constellated by cellular elements and a network of soluble constituents that contribute to tumor progression. In the multitude of the secreted molecules, the endothelin-1 (ET-1) has emerged to be implicated in the tumor/TME interplay; however, the molecular mechanisms induced by the ET-1-driven feed-forward loops (FFL) and associated with the HG-SOC metastatic potential need to be further investigated. The tracking of the patient-derived (PD) HG-SOC cell transcriptome by RNA-seq identified the vascular endothelial growth factor (VEGF) gene and its associated signature among those mostly up-regulated by ET-1 and down-modulated by the dual ET-1R antagonist macitentan. Within the ligand-receptor pairs concurrently expressed in PD-HG-SOC cells, endothelial cells and activated fibroblasts, we discovered two intertwined FFL, the ET-1/ET-1R and VEGF/VEGF receptors, concurrently activated by ET-1 and shutting-down by macitentan, or by the anti-VEGF antibody bevacizumab. In parallel, we observed that ET-1 fine-tuned the tumoral and stromal secretome toward a pro-invasive pattern. Into the fray of the HG-SOC/TME double and triple co-cultures, the secretion of ET-1 and VEGF, that share a common co-regulation, was inhibited upon the administration of macitentan. Functionally, macitentan, mimicking the effect of bevacizumab, interfered with the HG-SOC/TME FFL-driven communication that fuels the HG-SOC invasive behavior. The identification of ET-1 and VEGF FFL as tumor and TME actionable vulnerabilities, reveals how ET-1R blockade, targeting the HG-SOC cells and the TME simultaneously, may represent an effective therapeutic option for HG-SOC patients.
Collapse
Affiliation(s)
- Piera Tocci
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Celia Roman
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Sacconi
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Ivan Molineris
- Department of Life Science and System Biology, University of Turin, Turin, Italy
| | - Giovanni Tonon
- Center for Omics Sciences (COSR) and Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
5
|
Gilley P, Zhang K, Abdoli N, Sadri Y, Adhikari L, Fung KM, Qiu Y. Utilizing a Pathomics Biomarker to Predict the Effectiveness of Bevacizumab in Ovarian Cancer Treatment. Bioengineering (Basel) 2024; 11:678. [PMID: 39061760 PMCID: PMC11273783 DOI: 10.3390/bioengineering11070678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The purpose of this investigation is to develop and initially assess a quantitative image analysis scheme that utilizes histopathological images to predict the treatment effectiveness of bevacizumab therapy in ovarian cancer patients. As a widely accessible diagnostic tool, histopathological slides contain copious information regarding underlying tumor progression that is associated with tumor prognosis. However, this information cannot be readily identified by conventional visual examination. This study utilizes novel pathomics technology to quantify this meaningful information for treatment effectiveness prediction. Accordingly, a total of 9828 features were extracted from segmented tumor tissue, cell nuclei, and cell cytoplasm, which were categorized into geometric, intensity, texture, and subcellular structure features. Next, the best performing features were selected as the input for SVM (support vector machine)-based prediction models. These models were evaluated on an open dataset containing a total of 78 patients and 288 whole slides images. The results indicated that the sufficiently optimized, best-performing model yielded an area under the receiver operating characteristic (ROC) curve of 0.8312. When examining the best model's confusion matrix, 37 and 25 cases were correctly predicted as responders and non-responders, respectively, achieving an overall accuracy of 0.7848. This investigation initially validated the feasibility of utilizing pathomics techniques to predict tumor responses to chemotherapy at an early stage.
Collapse
Affiliation(s)
- Patrik Gilley
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA (N.A.)
| | - Ke Zhang
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA (N.A.)
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Neman Abdoli
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA (N.A.)
| | - Youkabed Sadri
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA (N.A.)
| | - Laura Adhikari
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA (K.-M.F.)
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA (K.-M.F.)
| | - Yuchen Qiu
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA (N.A.)
| |
Collapse
|
6
|
Calmon MS, Lemos FFB, Silva Luz M, Rocha Pinheiro SL, de Oliveira Silva LG, Correa Santos GL, Rocha GR, Freire de Melo F. Immune pathway through endometriosis to ovarian cancer. World J Clin Oncol 2024; 15:496-522. [PMID: 38689629 PMCID: PMC11056862 DOI: 10.5306/wjco.v15.i4.496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/22/2024] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disease, defined by the presence of functional endometrial tissue outside of the uterine cavity. This disease is one of the main gynecological diseases, affecting around 10%-15% women and girls of reproductive age, being a common gynecologic disorder. Although endometriosis is a benign disease, it shares several characteristics with invasive cancer. Studies support that it has been linked with an increased chance of developing endometrial ovarian cancer, representing an earlier stage of neoplastic processes. This is particularly true for women with clear cell carcinoma, low-grade serous carcinoma and endometrioid. However, the carcinogenic pathways between both pathologies remain poorly understood. Current studies suggest a connection between endometriosis and endometriosis-associated ovarian cancers (EAOCs) via pathways associated with oxidative stress, inflammation, and hyperestrogenism. This article aims to review current data on the molecular events linked to the development of EAOCs from endometriosis, specifically focusing on the complex relationship between the immune response to endometriosis and cancer, including the molecular mechanisms and their ramifications. Examining recent developments in immunotherapy and their potential to boost the effectiveness of future treatments.
Collapse
Affiliation(s)
- Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Reis Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
7
|
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
|
8
|
Karacin C, Sunar V, Urakci Z, Yilmaz A, Ayhan M, Ersoy M, Guven DC, Erturk I, Durmus Y, Karacin P, Boran N, Ustun YE, Meydan M, Dogan M, Oksuzoğlu B, Ates O, Karaca M, Uncu D, Ergun Y, Arik Z. Efficacy and safety of bevacizumab in patients with low-grade serous ovarian cancer. Future Oncol 2024; 20:207-214. [PMID: 38328890 DOI: 10.2217/fon-2023-0763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Aim: To investigate the efficacy and safety of bevacizumab in patients with recurrent low-grade serous ovarian carcinoma. Materials & methods: The data of patients who received at least two cycles of bevacizumab in combination with chemotherapy were retrospectively recorded. Results: The median age of 51 patients was 56 (range: 33-75) years. The complete response rate was 10.4% and the partial response rate was 43.7%. The objective response rate was 54.1%. Median progression-free survival was 15.9 months (95% CI: 9.1-22.6) and median overall survival was 42.5 months (95% CI: 37.2-47.8). Conclusion: Bevacizumab with chemotherapy is an effective option for treating recurrent ovarian low-grade serous carcinoma.
Collapse
Affiliation(s)
- Cengiz Karacin
- Department of Medical Oncology, University of Health Sciences, Dr Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Ankara, Turkey
| | - Veli Sunar
- Depatment of Medical Oncology, Losante Children & Adult Hospital, Ankara, Turkey
| | - Zuhat Urakci
- Departmen of Medical Oncology, Dicle University, Diyarbakir, Turkey
| | - Ali Yilmaz
- Department of Medical Oncology, Atatürk University Training Hospital, Erzurum, Turkey
| | - Murat Ayhan
- Department of Medical Oncology, Kartal Training & Research Hospital, Istanbul, Turkey
| | - Mustafa Ersoy
- Department of Medical Oncology, Osmangazi University, Eskisehir, Turkey
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Ismail Erturk
- Department of Medical Oncology, University of Health Sciences, Gulhane Training & Research Hospital, Ankara, Turkey
| | - Yasin Durmus
- Department of Gynecology, Etlik Zubeyde Hanim Training & Research Hospital, Ankara, Turkey
| | - Pinar Karacin
- Department of Gynecology, Etlik Zubeyde Hanim Training & Research Hospital, Ankara, Turkey
| | - Nurettin Boran
- Department of Gynecology, Etlik Zubeyde Hanim Training & Research Hospital, Ankara, Turkey
| | - Yaprak Engin Ustun
- Department of Gynecology, Etlik Zubeyde Hanim Training & Research Hospital, Ankara, Turkey
| | - Mutlu Meydan
- Department of Gynecology, Zekai Tahir Burak Training & Research Hospital, Ankara, Turkey
| | - Mutlu Dogan
- Department of Medical Oncology, University of Health Sciences, Dr Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Ankara, Turkey
| | - Berna Oksuzoğlu
- Department of Medical Oncology, University of Health Sciences, Dr Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Ankara, Turkey
| | - Ozturk Ates
- Department of Medical Oncology, University of Health Sciences, Dr Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Ankara, Turkey
| | - Mustafa Karaca
- Department of Medical Oncology, Antalya Training & Research Hospital, Antalya, Turkey
| | - Dogan Uncu
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Yakup Ergun
- Department of Medical Oncology, Antalya City Hospital, Antalya, Turkey
| | - Zafer Arik
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
9
|
Kang N, Fan B, Sun Y, Xue P, Liu Y. Novel specific anti-ESM1 antibodies overcome tumor bevacizumab resistance by suppressing angiogenesis and metastasis. Cancer Sci 2023; 114:4413-4425. [PMID: 37715566 PMCID: PMC10637069 DOI: 10.1111/cas.15939] [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: 03/31/2023] [Revised: 08/07/2023] [Accepted: 08/13/2023] [Indexed: 09/17/2023] Open
Abstract
Suppressing tumors through anti-angiogenesis has been established as an effective clinical treatment strategy. Bevacizumab, a monoclonal antibody, is commonly used in various indications. However, two major challenges limit the long-term efficacy of bevacizumab: drug resistance and side effects. Bevacizumab resistance has been extensively studied at the molecular level, but no drug candidates have been developed for clinical use to overcome this resistance. In a previous study conducted by our team, a major finding was that high expression of ESM1 in bevacizumab-resistant tumors is associated with an unfavorable response to treatment. In particular, an increase in ESM1 expression contributes to heightened lung metastasis and microvascular density, which ultimately decreases the tumor's sensitivity to bevacizumab. In contrast, the silencing of ESM1 results in reduced angiogenesis and suppressed tumor growth in tumors resistant to bevacizumab. We put forward the hypothesis that targeting ESM1 could serve as a therapeutic strategy in overcoming bevacizumab resistance. In this study, a variety of anti-ESM1 antibodies with high affinity to human ESM1 were successfully prepared and characterized. Our in vivo study confirmed the establishment of a bevacizumab-resistant human colorectal cancer model and further demonstrated that the addition of anti-ESM1 monoclonal antibodies to bevacizumab treatment significantly improved tumor response while downregulating DLL4 and MMP9. In conclusion, our study suggests that anti-hESM1 monoclonal antibodies have the potential to alleviate or overcome bevacizumab resistance, thereby providing new strategies and drug candidates for clinical research in the treatment of bevacizumab-resistant colorectal cancer.
Collapse
Affiliation(s)
- Nannan Kang
- School of Life Science & TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Buxi Fan
- School of Life Science & TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Yao Sun
- School of Life Science & TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Peilin Xue
- School of Life Science & TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Yu Liu
- School of Life Science & TechnologyChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
10
|
Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2023; 49:2563-2574. [PMID: 37565583 DOI: 10.1111/jog.15761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
AIM This review aimed to describe the potential for therapeutic targeting of the JAK/STAT signaling pathway by repurposing the clinically-approved JAK inhibitor ruxolitinib in the patients with epithelial ovarian cancer (OC) setting. METHODS We reviewed publications that focus on the inhibition of the JAK/STAT pathway in hematological and solid malignancies including OC. RESULTS Preclinical studies showed that ruxolitinib effectively reduces OC cell viability and metastasis and enhances the anti-tumor activity of chemotherapy drugs. There are a number of recent clinical trials exploring the role of JAK/STAT inhibition in solid cancers including OC. Early results have not adequately supported efficacy in solid tumors. However, there are preclinical data and clinical studies supporting the use of ruxolitinib in combination with both chemotherapy and other targeted drugs in OC setting. CONCLUSION Inflammatory conditions and persistent activation of the JAK/STAT pathway are associated with tumourigenesis and chemoresistance, and therapeutic blockade of this pathway shows promising results. For women with OC, clinical investigation exploring the role of ruxolitinib in combination with chemotherapy agents or other targeted therapeutics is warranted.
Collapse
Affiliation(s)
- Irfan Yunianto
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
- Department of Biology Education, Universitas Ahmad Dahlan, Indonesia
| | - Margaret Currie
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
11
|
Hou Z, Lan C, Huang X, Salcedo-Hernández RA, EL-Tawab S. Efficacy, safety and pharmacokinetics of apatinib plus etoposide versus apatinib alone for platinum-resistant recurrent ovarian cancer: protocol of a multicenter, open-label, randomized phase 2 trial. Transl Cancer Res 2023; 12:2959-2967. [PMID: 37969395 PMCID: PMC10643946 DOI: 10.21037/tcr-23-1924] [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: 08/23/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Background Currently preferred single-agent nonplatinum chemotherapy or its combination with bevacizumab results in a low response rate and modest survival benefit for platinum-resistant recurrent ovarian cancer, and thus more effective regimens are needed. In our previous phase 2 trial, apatinib plus etoposide showed promising efficacy and an acceptable safety profile in platinum-resistant recurrent ovarian cancer patients. Due to the single-arm design, the role of apatinib still needs to be determined. Methods In this phase 2 trial, 54 adult patients with platinum-resistant current ovarian cancer will be recruited at 17 sites in China. Patients with prior administration of small-molecule tyrosine kinase inhibitors or etoposide will be excluded. Patients will be randomized (1:1) to receive apatinib (375 mg, orally, once daily) alone or in combination with etoposide (50 mg, orally on days 1-14 of each 21-day cycle) until disease progression or intolerable toxicity. Randomization will be performed using a computerized central randomization system, stratified by platinum resistance for the first time (yes or no). Imaging examinations will be conducted every 6 weeks. The primary endpoint is the objective response rate (ORR) according to the Response Evaluation Criteria In Solid Tumors (version 1.1), which will be compared between groups using the Cochran-Mantel-Haenszel test. Discussion This study will provide prospective data of 2 experimental regimens using a randomized design. It will help determine whether apatinib monotherapy can provide favorable clinical benefits or needs to be combined with chemotherapy to be effective. Trial Registration ClinicalTrials.gov Identifier: NCT04383977. It was registered on May 12, 2020.
Collapse
Affiliation(s)
- Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Chunyan Lan
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Huang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rosa A. Salcedo-Hernández
- Gynecological Cancers Division, Instituto Nacional de Cancerología de México (Mexico’s National Cancer Institute), Mexico City, Mexico
| | - Sally EL-Tawab
- Oxford Gynaecological Cancer Centre, Churchill Hospital, Oxford University Hospitals Foundation Trust, Oxford, UK
| |
Collapse
|
12
|
Silva TS, Martins MR, Batista TL, Martins ED, Fernandes MH, Hinrichsen EA. Case Report: Abdominal Wall Reconstruction in a High-Risk Patient With Incisional Hernia and Complications From Oncological Treatment. JOURNAL OF ABDOMINAL WALL SURGERY : JAWS 2023; 2:11767. [PMID: 38312430 PMCID: PMC10831638 DOI: 10.3389/jaws.2023.11767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/30/2023] [Indexed: 02/06/2024]
Abstract
Introduction: A high risk patient with evisceration underwent to abdominal wall reconstruction without mesh or drains. We present a case of a 62 years-old female patient with a significant medical history of Wilson's disease-related hepatopathy Child-Pugh class B classification, sequelae of a stroke, and relevant surgical background including total hysterectomy, oophorectomy, and Hartmann's procedure for ovarian neoplasm stage 3. The patient developed a large incisional hernia in the midline incision while undergoing Bevacizumab (Avastin) treatment for clinical oncology. During an attempt at skin closure due to erosion and necrosis, there was progressive deterioration leading to evisceration. We opted for abdominal wall reconstruction by transposing the hernia sac without using mesh and employing hemostatic powder (Arista) to mitigate the risk of bleeding in a high-risk patient due to recent bevacizumab use and hepatopathy. The patient had a favorable postoperative course without any other intervention in abdominal wall. Patient developed worsening hepatic function with the presence of ascites, constipation, and disorientation. On the 6th day postoperative, a tomography was performed, which showed colonic distension without obstructive factors and a slight amount of supra-aponeurotic fluid. The patient was discharged on the 10th day postoperative after improvement of the condition with clinical treatment. The patient has been progressing under outpatient follow-up for 5 months, with a resumption of chemotherapy cycles and no evidence of hernia recurrence. Conclusion: Further studies and long-term follow-up are necessary to evaluate the efficacy and safety of hernia sac transposition as a mesh-free technique and the use of hemostatic powder without drains in high-risk patients. However, our case highlights the potential feasibility of these approaches in carefully selected cases.
Collapse
Affiliation(s)
- Thiago Souza Silva
- Real Hospital Português, Recife, Brazil
- European Hernia Society (EHS), Gdansk, Poland
| | | | | | - Euclides Dias Martins
- Real Hospital Português, Recife, Brazil
- Colégio Brasileiro de Cirurgiões, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
13
|
Cartmel B, Li F, Zhou Y, Gottlieb L, Lu L, Mszar R, Harrigan M, Ligibel JA, Gogoi R, Schwartz PE, Risch HA, Irwin ML. Randomized trial of exercise on cancer-related blood biomarkers and survival in women with ovarian cancer. Cancer Med 2023; 12:15492-15503. [PMID: 37269192 PMCID: PMC10417064 DOI: 10.1002/cam4.6187] [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: 10/13/2022] [Revised: 04/13/2023] [Accepted: 05/21/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND In randomized trials in women with breast cancer, exercise has been shown to have beneficial effects on cancer-related circulating biomarkers that may impact survival. Such studies are lacking for ovarian cancer. METHODS This secondary analysis of a published randomized controlled trial examined the impact of a 6-month exercise intervention versus attention-control on change in prespecified circulating biomarkers (cancer antigen 125 (CA-125), C-reactive protein (CRP), insulin-like growth factor-1(IGF-1), insulin and leptin) in a subset of participants who provided a fasting blood draw (N = 104/144) at enrollment and at 6 months. Change in biomarkers between study arms was compared using a linear mixed effects model analysis. An exploratory analysis of the exercise intervention versus attention-control on all-cause mortality included all (N = 144) participants. All statistical tests were two-sided. RESULTS Participants included in the biomarker analysis were 57.0 ± 8.8 (mean ± SD) years old and 1.6 ± 0.9 years post-diagnosis. Adherence to the exercise intervention was 176.4 ± 63.5 min/week. Post intervention IGF-1 (group difference in change: -14.2 (-26.1 to -2.3) ng/mL (least squared means (95% CI))) and leptin (-8.9 (-16.5 to -1.4) ng/mL) were significantly reduced in the exercise group (N = 53) compared to those in attention-control (N = 51). No group difference in change was seen for CA-125 (p = 0.54), CRP (p = 0.95), or insulin (p = 0.37). With median follow-up of 70 months [range 6.6-105.4 months], 50/144 (34.7%) (exercise group; 24/74 (32.4%) versus attention-control group; 26/70 (37.1%)) participants died with no between group difference in overall survival (p = 0.99). CONCLUSIONS Further studies are needed to determine the clinical significance of exercise-induced changes in cancer-related circulating biomarkers in women with ovarian cancer.
Collapse
Affiliation(s)
- Brenda Cartmel
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Fang‐yong Li
- Department of BiostatisticsYale School of Public HealthNew HavenConnecticutUSA
| | - Yang Zhou
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Linda Gottlieb
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| | - Lingeng Lu
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Reed Mszar
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| | - Maura Harrigan
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| | - Jennifer A. Ligibel
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Radhika Gogoi
- Gynecologic Oncology, Women and Children's InstituteGeisinger Health SystemDanvillePennsylvaniaUSA
- Present address:
Department of Obstetrics & GynecologyWayne State UniversityDetroitMichiganUSA
| | - Peter E. Schwartz
- Yale Cancer CenterNew HavenConnecticutUSA
- Section of Medical Oncology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Harvey A. Risch
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Melinda L. Irwin
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| |
Collapse
|
14
|
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: 4.0] [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
|
15
|
Choi MS, Kim JH, Lee CY, Lee YM, Lee S, Chang HK, Kim HJ, Heo K. Gentian Violet Inhibits Cell Proliferation through Induction of Apoptosis in Ovarian Cancer Cells. Biomedicines 2023; 11:1657. [PMID: 37371752 DOI: 10.3390/biomedicines11061657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Gentian violet (GV) is known to have antibacterial and antifungal effects, but recent studies have demonstrated its inhibitory effects on the growth of several types of cancer cells. Here, we investigated the anticancer efficacy of GV in ovarian cancer cells. GV significantly reduced the proliferation of OVCAR8, SKOV3, and A2780 cells. Results of transferase dUTP nick and labeling (TUNEL) assay and Western blot assay indicated that the inhibitory effect of GV on ovarian cancer cells was due to the induction of apoptosis. Moreover, GV significantly increased reactive oxygen species (ROS) and upregulated the expression of p53, PUMA, BAX, and p21, critical components for apoptosis induction, in ovarian cancer cells. Our results suggest that GV is a novel antiproliferative agent and is worthy of exploration as a potential therapeutic agent for ovarian cancer.
Collapse
Affiliation(s)
- Min Sung Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Hyeon Kim
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Chae Yeon Lee
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Yul Min Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Kyun Chang
- Department of Obstetrics and Gynecology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15855, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
16
|
Choi YN, Seo TW, Lee YT, Jeong DH, Yoo SJ. Nuclear endonuclease G controls cell proliferation in ovarian cancer. FEBS Open Bio 2023; 13:655-669. [PMID: 36734593 PMCID: PMC10068316 DOI: 10.1002/2211-5463.13572] [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: 08/26/2022] [Revised: 12/22/2022] [Accepted: 02/02/2023] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is characterized by a high degree of genetic heterogeneity. Platinum-based chemotherapy and some gene-targeted therapies have shown limited treatment efficacy due to toxicity and recurrence, and thus, it is essential to identify additional therapeutic targets based on an understanding of the pathological mechanism. Here, we report that endonuclease G, which exhibits altered expression in ovarian cancer, does not function as a cell death effector that digests chromosomal DNA in ovarian cancer. Endonuclease G is modulated by intracellular reactive oxygen species dynamics and plays a role in cell proliferation in ovarian cancer, suggesting that targeting endonuclease G alone or in combination with other antitumor agents may have the potential for development into a treatment for endonuclease G-overexpressing cancers, including ovarian cancer.
Collapse
Affiliation(s)
- Ye Na Choi
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Tae Woong Seo
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Yui Taek Lee
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Dar Heum Jeong
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Soon Ji Yoo
- Department of Biology, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
17
|
Trifanescu OG, Gales LN, Tanase BC, Marinescu SA, Trifanescu RA, Gruia IM, Paun MA, Rebegea L, Mitrica R, Serbanescu L, Anghel RM. Prognostic Role of Vascular Endothelial Growth Factor and Correlation with Oxidative Stress Markers in Locally Advanced and Metastatic Ovarian Cancer Patients. Diagnostics (Basel) 2023; 13:diagnostics13010166. [PMID: 36611458 PMCID: PMC9818969 DOI: 10.3390/diagnostics13010166] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) plays an important role in tumor progression in ovarian cancer, but the complex mechanism and interaction with oxidative stress are not fully understood. METHODS A prospective study included 52 patients with ovarian adenocarcinoma stage IIIA-IV. Serum VEGF and reactive oxygen species (ROS) such as malondialdehyde and ceruloplasmin were measured. RESULTS VEGF levels were elevated (mean 1014.7 ± 165 pg/mL), especially in patients with macroscopic residual disease (1058 vs. 810 pg/mL, p = 0.0001). Median progression-free survival (PFS) and overall survival (OS) were 6 and 40 months in patients with a very high VEGF (over 1200 pg/mL), 11 and 48 months in patients with VEGF between 1000-1200 pg/mL, 18 and 84 months in patients with VEGF between 800-1000 pg/mL, and not reached in patients with normal VEGF. Increased VEGF values were associated with a 2.6-fold increased risk of disease progression (HR = 2.60, 95% CI 1.69-3.99), and a 1.4-fold increased risk of death (HR = 1.4, 95% CI 1.15-1.91, p = 0.002). Receiver operator characteristic (ROC) curves were used to validate VEGF as a prognostic factor and the area under the curve (AUC) was 0.814, p = 0.036 for PFS and 0.729, p = 0.043, for OS. There was a positive correlation between VEGF and malondialdehyde, Pearson coefficient of 0.35, p = 0.0001. CONCLUSIONS VEGF and malondialdehyde are important prognostic markers in ovarian cancer, especially in macroscopic residual disease, and there is a positive correlation between angiogenesis and oxidative stress.
Collapse
Affiliation(s)
- Oana Gabriela Trifanescu
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Laurentia Nicoleta Gales
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
- Correspondence: (L.N.G.); (R.A.T.)
| | - Bogdan Cosmin Tanase
- Thoracic Surgery, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Serban Andrei Marinescu
- Oncologic Surgery I, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Raluca Alexandra Trifanescu
- “C. I. Parhon” Institute of Endocrinology, 011863 Bucharest, Romania
- Department of Endocrinology C. I. Parhon, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Correspondence: (L.N.G.); (R.A.T.)
| | - Iuliana Maria Gruia
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Mihai Andrei Paun
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Laura Rebegea
- Medical Clinical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galați, Romania
| | - Radu Mitrica
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Luiza Serbanescu
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| |
Collapse
|
18
|
Dey A, Mitra A, Pathak S, Prasad S, Zhang AS, Zhang H, Sun XF, Banerjee A. Recent Advancements, Limitations, and Future Perspectives of the use of Personalized Medicine in Treatment of Colon Cancer. Technol Cancer Res Treat 2023; 22:15330338231178403. [PMID: 37248615 PMCID: PMC10240881 DOI: 10.1177/15330338231178403] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/18/2023] [Accepted: 03/13/2023] [Indexed: 08/29/2024] Open
Abstract
Due to the heterogeneity of colon cancer, surgery, chemotherapy, and radiation are ineffective in all cases. The genomic profile and biomarkers associated with the process are considered in personalized medicine, along with the patient's personal history. It is based on the response of the targeted therapies to specific genetic variations. The patient's genetic transcriptomic and epigenetic features are evaluated, and the best therapeutic approach and diagnostic testing are identified through personalized medicine. This review aims to summarize all the necessary, updated information on colon cancer related to personalized medicine. Personalized medicine is gaining prominence as generalized treatments are finding it challenging to contain colon cancer cases which currently rank fourth among global cancer incidence while being the fifth largest in total death cases worldwide. In personalized therapy, patients are grouped into specific categories, and the best therapeutic approach is chosen based on evaluating their molecular features. Various personalized strategies are currently being explored in the treatment of colon cancer involving immunotherapy, phytochemicals, and other biomarker-specific targeted therapies. However, significant challenges must be overcome to integrate personalized medicine into healthcare systems completely. We look at the various signaling pathways and genetic and epigenetic alterations associated with colon cancer to understand and identify biomarkers useful in targeted therapy. The current personalized therapies available in colon cancer treatment and the strategies being explored to improve the existing methods are discussed. This review highlights the advantages and limitations of personalized medicine in colon cancer therapy. The current scenario of personalized medicine in developed countries and the challenges faced in middle- and low-income countries are also summarized. Finally, we discuss the future perspectives of personalized medicine in colon cancer and how it could be integrated into the healthcare systems.
Collapse
Affiliation(s)
- Amit Dey
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Abhijit Mitra
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Suhanya Prasad
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Białystok, Poland
| | | | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Orebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| |
Collapse
|