1
|
Patel D, Dickson AL, Zickuhr GM, Um IH, Read OJ, Czekster CM, Mullen P, Harrison DJ, Bré J. Defining the mode of action of cisplatin combined with NUC-1031, a phosphoramidate modification of gemcitabine. Transl Oncol 2024; 50:102114. [PMID: 39299019 PMCID: PMC11426158 DOI: 10.1016/j.tranon.2024.102114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
The combination of gemcitabine with platinum agents is a widely used chemotherapy regimen for a number of tumour types. Gemcitabine plus cisplatin remains the current therapeutic choice for biliary tract cancer. Gemcitabine is associated with multiple cellular drug resistance mechanisms and other limitations and has thereforelined in use. NUC-1031 (Acelarin) is a phosphorylated form of gemcitabine, protected by the addition of a phosphoramidate moiety, developed to circumvent the key limitations and generate high levels of the cytotoxic metabolite, dFdCTP. The rationale for combination of gemcitabine and cisplatin is determined by in vitro cytotoxicity. This, however, does not offer an explanation of how these drugs lead to cell death. In this study we investigate the mechanism of action for NUC-1031 combined with cisplatin as a rationale for treatment. NUC-1031 is metabolised to dFdCTP, detectable up to 72 h post-treatment and incorporated into DNA, to stall the cell cycle and cause DNA damage in biliary tract and ovarian cancer cell lines. In combination with cisplatin, DNA damage was increased and occurred earlier compared to monotherapy. The damage associated with NUC-1031 may be potentiated by a second mechanism, via binding the RRM1 subunit of ribonucleotide reductase and perturbing the nucleotide pools; however, this may be mitigated by increased RRM1 expression. The implication of this was investigated in case studies from a Phase I clinical trial to observe whether baseline RRM1 expression in tumour tissue at time of diagnosis correlates with patient survival.
Collapse
Affiliation(s)
- Dillum Patel
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK.
| | - Alison L Dickson
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Greice M Zickuhr
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - In Hwa Um
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - Oliver J Read
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Clarissa M Czekster
- School of Biology, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - Peter Mullen
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK
| | - David J Harrison
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Jennifer Bré
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, UK; NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| |
Collapse
|
2
|
He W, Zhu H, Zhang S, Shu G, Lei H, Wang M, Yin G, Ni X, Wu Q. Epigenetic editing of BRCA1 promoter increases cisplatin and olaparib sensitivity of ovarian cancer cells. Epigenetics 2024; 19:2357518. [PMID: 38796857 PMCID: PMC11135871 DOI: 10.1080/15592294.2024.2357518] [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: 01/24/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Drug resistance is the primary contributor to the high mortality rate of ovarian cancer (OC). The loss of BRCA1/2 function is linked to drug sensitivity in OC cells. The aim of this study is to enhance the drug sensitivity of OC cells by inducing BRCA1 dysfunction through promoter epigenetic editing. Epigenetic regulatory regions within the BRCA1 promoter, affecting gene expression, were initially discerned through analysis of clinical samples. Subsequently, we designed and rigorously validated epigenetic editing tools. Ultimately, we evaluated the cisplatin and olaparib sensitivity of the OC cells after editing. The BRCA1 promoter contains two CpG-rich regions, with methylation of the region covering the transcription start site (TSS) strongly correlating with transcription and influencing OC development, prognosis, and homologous recombination (HR) defects. Targeting this region in OC cells using our designed epigenetic editing tools led to substantial and persistent DNA methylation changes, accompanied by significant reductions in H3K27ac histone modifications. This resulted in a notable suppression of BRCA1 expression and a decrease in HR repair capacity. Consequently, edited OC cells exhibited heightened sensitivity to cisplatin and olaparib, leading to increased apoptosis rates. Epigenetic inactivation of the BRCA1 promoter can enhance cisplatin and olaparib sensitivity of OC cells through a reduction in HR repair capacity, indicating the potential utility of epigenetic editing technology in sensitization therapy for OC.
Collapse
Affiliation(s)
- Wanhong He
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Haijun Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Sufen Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Han Lei
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xiaohua Ni
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| |
Collapse
|
3
|
Jackson-Spence F, Ackerman C, Jones R, Toms C, Jovaisaite A, Young M, Hussain S, Protheroe A, Birtle A, Chakraborti P, Huddart R, Jagdev S, Bahl A, Sundar S, Crabb S, Powles T, Szabados B. Biomarkers associated with survival in patients with platinum-refractory urothelial carcinoma treated with paclitaxel. Urol Oncol 2024; 42:372.e1-372.e10. [PMID: 39025719 DOI: 10.1016/j.urolonc.2024.05.015] [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: 09/15/2023] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Taxane- based chemotherapy is widely used in patients with platinum- and immunotherapy refractory, metastatic urothelial carcinoma (mUC). Outcomes are poor and biomarkers associated with outcome are lacking. We aim to identify cancer hallmarks associated with survival in patients receiving paclitaxel. METHODS Whole-transcriptome profiles were generated for a subset of patients enrolled in a randomised phase II study investigating paclitaxel and pazopanib in platinum refractory mUC (PLUTO, EudraCT 2011-001841-34). Estimates of gene expression were calculated and input into the Almac proprietary analysis pipeline and signature scores were calculated using ClaraT V3.0.0. Ten key gene signatures were assessed: Immuno-Oncology, Epithelial to Mesenchymal Transition, Angiogenesis, Proliferation, Cell Death, Genome Instability, Energetics, Inflammation, Immortality and Evading Growth. Hazard ratios were calculated using Cox regression model and Kaplan-Meier methods were used to estimate progression free survival (PFS) and overall survival (OS). RESULTS 38 and 45 patients treated with paclitaxel or pazopanib were included. Patients with high genome instability expression treated with paclitaxel had significantly improved survival with a HR of 0.29 (95% CI: 0.14-0.61, p=0.001) and HR 0.34 (95% CI: 0.17-0.69, p=0.003) for PFS and OS, respectively. Similarly, patients with high evading growth suppressor expression treated with paclitaxel had improved PFS and OS with a HR of 0.35 (95% CI: 0.19-0.77, p=0.007) and HR 0.46 (95% CI: 0.23-0.91, p=0.026), respectively. No other gene signatures had significant impact on outcome. In both paclitaxel and pazopanib cohorts, angiogenesis activation was associated with worse PFS and OS, and VEGF targeted therapy did not improve outcomes. CONCLUSION High Genome-instability and Evading-growth suppressor biologies are associated with improved survival in patients with platinum refractory mUC receiving paclitaxel. These may refine mUC risk stratification and guide treatment decision in the future.
Collapse
Affiliation(s)
| | - Charlotte Ackerman
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Robert Jones
- Department of Genitourinary Oncology, University of Glasgow, Glasgow, Scotland, UK
| | - Charlotte Toms
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Agne Jovaisaite
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Matthew Young
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Syed Hussain
- Department of Genitourinary Oncology, University of Liverpool, Liverpool, UK
| | - Andrew Protheroe
- Department of Genitourinary Oncology, Churchill Hospital, Oxford, UK
| | - Alison Birtle
- Department of Genitourinary Oncology, Preston Hospital, Preston, UK
| | - Prabir Chakraborti
- Department of Genitourinary Oncology, Derby Hospitals NHS Foundation trust, Derby, UK
| | - Robert Huddart
- Department of Genitourinary Oncology, Institute of Cancer Research, Sutton, UK
| | - Santinder Jagdev
- Department of Genitourinary Oncology, St James's University Hospital, Leeds, UK
| | - Amit Bahl
- Department of Genitourinary Oncology, Bristol Haematology and Oncology Centre, Bristol, UK
| | - Santhanam Sundar
- Department of Genitourinary Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Simon Crabb
- Department of Genitourinary Oncology, University of Southampton, Southampton UK
| | - Thomas Powles
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK.
| | - Bernadett Szabados
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| |
Collapse
|
4
|
de Menezes Pereira G, Bormio Nunes JH, Macedo VS, Pereira DH, Buglio KE, Affonso DD, Ruiz ALTG, de Carvalho JE, Frajácomo SCL, Lustri WR, Lima CSP, Bergamini FRG, Cuin A, Masciocchi N, Corbi PP. Antibacterial profile and antiproliferative activities over human tumor cells of new silver(I) complexes containing two distinct trifluoromethyl uracil isomers. J Inorg Biochem 2024; 262:112752. [PMID: 39366100 DOI: 10.1016/j.jinorgbio.2024.112752] [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: 09/01/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
New silver(I) complexes of 5-(trifluoromethyl)uracil (5TFMU) and 6-(trifluoromethyl)uracil (6TFMU) isomers were synthesized, characterized, and evaluated as antibacterial and antiproliferative agents. Based on elemental and thermogravimetric analyses, the Ag-5TFMU and Ag-6TFMU species are formulated as AgC5H2F3N2O2 and Ag2C5HF3N2O2, respectively. Infrared and 13C solid-state nuclear magnetic resonance spectroscopies suggest coordination of the trifluoromethyluracil isomers to silver by both nitrogen and oxygen atoms. Confirmation of their structure and connectivity was achieved, in the absence of single crystals of suitable quality, by state-of-the-art structural powder diffraction methods. In Ag-5TFMU, the organic ligand is tridentate and two distinct metal coordination environments are found (linear AgN2 as well as C2v AgO4 geometries), whereas Ag-6TFMU contains a complex polymeric structure with tetradentate dianionic 6TFMU moieties and five distinct AgX2 (X = N, O) fragments, further stabilized by ancillary (longer) Ag…O contacts. These species presented modest activity over Gram-positive and Gram-negative bacterial strains, whereas Ag-6TFMU was active over a set of tumor cells, with the best activity over prostate (PC-3) and kidney cell lines and selectivity indices of 4.6 and 1.3, respectively. On the other hand, Ag-5TFMU was active over all considered tumor cells except MCF-7 (breast cancer). The best activity was found for PC-3 cells, but no selectivity was observed. The Ag-5TFMU and Ag-6TFMU species also reduced the proliferation of tongue squamous cell carcinoma cell lines SCC - 4 and SCC-15. Preliminary biophysical assays by circular dichroism suggest that the Ag-5TFMU complex interacts with DNA by intercalation, an effect not seen in Ag-6TFMU.
Collapse
Affiliation(s)
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Vinicius Souza Macedo
- Chemistry Collegiate, Federal University of Tocantins - UFT, PO Box 66, 77402-970 Gurupi, TO, Brazil
| | - Douglas Henrique Pereira
- Chemistry Collegiate, Federal University of Tocantins - UFT, PO Box 66, 77402-970 Gurupi, TO, Brazil; Department of Chemistry, Technological Institute of Aeronautics (ITA), 12228-900 São José dos Campos, SP, Brazil
| | - Kaio Eduardo Buglio
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP, 13083-871 Campinas, SP, Brazil
| | - Daniele D Affonso
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP, 13083-871 Campinas, SP, Brazil
| | - Ana Lucia T G Ruiz
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP, 13083-871 Campinas, SP, Brazil
| | - João Ernesto de Carvalho
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP, 13083-871 Campinas, SP, Brazil
| | - Silmara Cristina L Frajácomo
- Department of Biological and Health Sciences, University of Araraquara - UNIARA, 14801-320 Araraquara, SP, Brazil
| | - Wilton R Lustri
- Department of Biological and Health Sciences, University of Araraquara - UNIARA, 14801-320 Araraquara, SP, Brazil
| | - Carmen Silvia Passos Lima
- Department of Anesthesiology, Oncology and Radiology, Faculty of Medical Sciences, University of Campinas - UNICAMP, Campinas 13083-887, SP, Brazil
| | - Fernando R G Bergamini
- Laboratory of Synthesis of Bioinspired Molecules, Institute of Chemistry, Federal University of Uberlândia - UFU, 38400-902 Uberlândia, MG, Brazil.
| | - Alexandre Cuin
- Institute of Exact Sciences, Department of Chemistry, Federal University of Juiz de Fora - UFJF, 36036-330 Juiz de Fora, MG, Brazil
| | - Norberto Masciocchi
- Department of Science and High Technology, University of Insubria, 21100 Varese, VA, Italy
| | - Pedro Paulo Corbi
- Institute of Chemistry, University of Campinas - UNICAMP, PO Box 6154, 13083-970 Campinas, SP, Brazil.
| |
Collapse
|
5
|
Krishna BM, Ramisetty SK, Garg P, Mohanty A, Wang E, Horne D, Awasthi S, Kulkarni P, Salgia R, Singhal SS. Enhancing carboplatin sensitivity in ovarian cancer cells by blocking the mercapturic acid pathway transporter. Carcinogenesis 2024; 45:696-707. [PMID: 39051454 DOI: 10.1093/carcin/bgae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/23/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Ral-binding/interacting protein (RLIP) acts as a transporter that responds to stress and provides protection, specifically against glutathione-electrophile conjugates and xenobiotic toxins. Its increased presence in malignant cells, especially in cancer, emphasizes its crucial antiapoptotic function. This is achieved by selectively regulating the cellular levels of proapoptotic oxidized lipid byproducts. Suppressing the progression of tumors in human xenografts can be achieved by effectively inhibiting RLIP, a transporter in the mercapturic acid pathway, without involving chemotherapy. Utilizing ovarian cancer (OC) cell lines (MDAH2774, OVCAR4, and OVCAR8), we observed that agents targeting RLIP, such as RLIP antisense and RLIP antibodies, not only substantially impeded the viability of OC cells but also remarkably increased their sensitivity to carboplatin. To delve further into the cytotoxic synergy between RLIP antisense, RLIP antibodies, and carboplatin, we conducted investigations in both cell culture and xenografts of OC cells. The outcomes revealed that RLIP depletion via phosphorothioate antisense led to rapid and sustained remissions in established subcutaneous human ovary xenografts. Furthermore, RLIP inhibition by RLIP antibodies exhibited comparable efficacy to antisense and enhanced the effectiveness of carboplatin in MDAH2774 OC xenografts. These investigations underscore RLIP as a central carrier crucial for supporting the survival of cancer cells, positioning it as a suitable focus for cancer treatment.
Collapse
Affiliation(s)
- B Madhu Krishna
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Sravani K Ramisetty
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Atish Mohanty
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Edward Wang
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Sanjay Awasthi
- Cayman Health, CTMH Doctors Hospital in Cayman Islands, George Town, Grand Cayman
| | - Prakash Kulkarni
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Ravi Salgia
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| | - Sharad S Singhal
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, United States
| |
Collapse
|
6
|
Thaklaewphan P, Wikan N, Potikanond S, Nimlamool W. Oxyresveratrol Enhances the Anti-Cancer Effect of Cisplatin against Epithelial Ovarian Cancer Cells through Suppressing the Activation of Protein Kinase B (AKT). Biomolecules 2024; 14:1140. [PMID: 39334906 PMCID: PMC11430010 DOI: 10.3390/biom14091140] [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: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Epithelial ovarian carcinoma poses a significant challenge due to its resistance to chemotherapy and propensity for metastasis, thereby reducing the effectiveness of conventional treatments. Hence, the identification of novel compounds capable of augmenting the anti-cancer efficacy of platinum-based chemotherapy is imperative. Oxyresveratrol (OXY), a derivative of resveratrol, has been demonstrated to possess antiproliferative and apoptosis-inducing effects across various cancer cell lines. Notably, OXY appears to exert its effects by inhibiting the PI3K/AKT/mTOR signaling pathway. However, the synergistic potential of OXY in combination with cisplatin against epithelial ovarian cancer has not yet been elucidated. The current study investigated the synergistic effects of OXY and cisplatin on the ovarian cancer cell lines SKOV3 and TOV21G. We found that OXY significantly enhanced cisplatin's ability to reduce cell viability, induce apoptosis, induce cell cycle arrest, and increase the proportion of cells in the sub-G1 phase. Furthermore, OXY treatment alone dose-dependently inhibited the production of anti-apoptotic proteins including Mcl-1, Bcl-xL, and XIAP under EGF activation. Mechanistically, OXY suppressed the PI3K/AKT/mTOR signaling pathway by reducing phosphorylated AKT, while having no discernible effect on the MAPK pathway. These findings highlight OXY's potential to enhance ovarian cancer cell sensitivity to chemotherapy, suggesting its development as a pharmaceutical adjunct for clinical use in combination therapies.
Collapse
Affiliation(s)
- Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
- Lanna Rice Research Center, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
7
|
Gogola-Mruk J, Pietrus M, Piechowicz M, Milian-Ciesielska K, Głód P, Wolnicka-Glubisz A, Szpor J, Ptak A. Low androgen/progesterone or high oestrogen/androgen receptors ratio in serous ovarian cancer predicts longer survival. Reprod Biol 2024; 24:100917. [PMID: 38970978 DOI: 10.1016/j.repbio.2024.100917] [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: 04/05/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 07/08/2024]
Abstract
The treatment of ovarian cancer (OC) remains one of the greatest challenges in gynaecological oncology. The presence of classic steroid receptors in OC makes hormone therapy an attractive option; however, the response of OC to hormone therapy is modest. Here, we compared the expression patterns of progesterone (PGR), androgen (AR) and oestrogen alpha (ERα) receptors between serous OC cell lines and non-cancer ovarian cells. These data were analysed in relation to steroid receptor expression profiles from patient tumour samples and survival outcomes using a bioinformatics approach. The results showed that ERα, PGR and AR were co-expressed in OC cell lines, and patient samples from high-grade and low-grade OC co-expressed at least two steroid receptors. High AR expression was negatively correlated, whereas ERα and PGR expression was positively correlated with patient survival. AR showed the opposite expression pattern to that of ERα and PGR in type 1 (SKOV-3) and 2 (OVCAR-3) OC cell lines compared with non-cancer (HOSEpiC) ovarian cells, with AR downregulated in type 1 and upregulated in type 2 OC. A low AR/PGR ratio and a high ESR1/AR ratio were associated with favourable survival outcomes in OC compared with other receptor ratios. Although the results must be interpreted with caution because of the small number of primary tumour samples analysed, they nevertheless suggest that the evaluation of ERα, AR and PGR by immunohistochemistry should be performed in patient biological material to plan future clinical trials.
Collapse
Affiliation(s)
- Justyna Gogola-Mruk
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow 30-387, Poland
| | - Miłosz Pietrus
- Department of Gynecology and Oncology, Faculty of Medicine, Jagiellonian University Medical College, Kraków 31-501, Poland
| | - Maryla Piechowicz
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow 30-387, Poland
| | - Katarzyna Milian-Ciesielska
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Krakow 31-531, Poland
| | - Paulina Głód
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow 30-387, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kracow 30-348, Poland
| | - Agnieszka Wolnicka-Glubisz
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow 30-387, Poland
| | - Joanna Szpor
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Krakow 31-531, Poland
| | - Anna Ptak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow 30-387, Poland.
| |
Collapse
|
8
|
Chen B, Liu J. Advances in ovarian tumor stem cells and therapy. Cell Biochem Biophys 2024; 82:1871-1892. [PMID: 38955927 DOI: 10.1007/s12013-024-01385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer is considered the most lethal among all gynecological malignancies due to its early metastatic dissemination, extensive spread, and malignant ascites. The current standard of care for advanced ovarian cancer involves a combination of cytoreductive surgery and chemotherapy utilizing platinum-based and taxane-based agents. Although initial treatment yields clinical remission in 70-80% of patients, the majority eventually develop treatment resistance and tumor recurrence. A growing body of evidence indicates the existence of cancer stem cells within diverse solid tumors, including ovarian cancer, which function as a subpopulation to propel tumor growth and disease advancement by means of drug resistance, recurrence, and metastasis. The presence of ovarian cancer stem cells is widely considered to be a significant contributor to the unfavorable clinical outcomes observed in patients with ovarian cancer, as they play a crucial role in mediating chemotherapy resistance, recurrence, and metastasis. Ovarian cancer stem cells possess the capacity to reassemble within the entirety of the tumor following conventional treatment, thereby instigating the recurrence of ovarian cancer and inducing resistance to treatment. Consequently, the creation of therapeutic approaches aimed at eliminating ovarian cancer stem cells holds great potential for the management of ovarian cancer. These cells are regarded as one of the most auspicious targets and mechanisms for the treatment of ovarian cancer. There is a pressing need for a comprehensive comprehension of the fundamental mechanisms of ovarian cancer's recurrence, metastasis, and drug resistance, alongside the development of effective strategies to overcome chemoresistance, metastasis, and recurrence. The implementation of cancer stem cell therapies may potentially augment the tumor cells' sensitivity to existing chemotherapy protocols, thereby mitigating the risks of tumor metastasis and recurrence, and ultimately improving the survival rates of ovarian cancer patients.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | - Jiaqi Liu
- Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
9
|
Tonon G, Rizzolio F, Visentin F, Scattolin T. Antibody Drug Conjugates for Cancer Therapy: From Metallodrugs to Nature-Inspired Payloads. Int J Mol Sci 2024; 25:8651. [PMID: 39201338 PMCID: PMC11355040 DOI: 10.3390/ijms25168651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
This review highlights significant advancements in antibody-drug conjugates (ADCs) equipped with metal-based and nature-inspired payloads, focusing on synthetic strategies for antibody conjugation. Traditional methods such us maleimide and succinimide conjugation and classical condensation reactions are prevalent for metallodrugs and natural compounds. However, emerging non-conventional strategies such as photoconjugation are gaining traction due to their milder conditions and, in an aspect which minimizes side reactions, selective formation of ADC. The review also summarizes the therapeutic and diagnostic properties of these ADCs, highlighting their enhanced selectivity and reduced side effects in cancer treatment compared to non-conjugated payloads. ADCs combine the specificity of monoclonal antibodies with the cytotoxicity of chemotherapy drugs, offering a targeted approach to the elimination of cancer cells while sparing healthy tissues. This targeted mechanism has demonstrated impressive clinical efficacy in various malignancies. Key future advancements include improved linker technology for enhanced stability and controlled release of cytotoxic agents, incorporation of novel, more potent, cytotoxic agents, and the identification of new cancer-specific antigens through genomic and proteomic technologies. ADCs are also expected to play a crucial role in combination therapies with immune checkpoint inhibitors, CAR-T cells, and small molecule inhibitors, leading to more durable and potentially curative outcomes. Ongoing research and clinical trials are expanding their capabilities, paving the way for more effective, safer, and personalized treatments, positioning ADCs as a cornerstone of modern medicine and offering new hope to patients.
Collapse
Affiliation(s)
- Giovanni Tonon
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano, Italy
| | - Fabiano Visentin
- Department of Molecular Sciences and Nanosystems, Università Ca’ Foscari Campus Scientifico, Via Torino 155, 30174 Venezia-Mestre, Italy; (G.T.); (F.R.)
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
10
|
Kim HJ, Kim LK, Kim A, Htwe KM, Heo TH, Shin KJ, Kim HJ, Yoon KD. IL-6 Inhibitory Compounds from the Aerial Parts of Piper attenuatum and Their Anticancer Activities on Ovarian Cancer Cell Lines. Molecules 2024; 29:2981. [PMID: 38998933 PMCID: PMC11242996 DOI: 10.3390/molecules29132981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Piper attenuatum Buch-Ham, a perennial woody vine belonging to the Piperaceae family, is traditionally used in Southeast Asia for treating various ailments such as malaria, headache, and hepatitis. This study described the isolation and identification of three new compounds, piperamides I-III (1-3), which belong to the maleimide-type alkaloid skeletons, along with fifteen known compounds (4-18) from the methanol extract of the aerial parts of P. attnuatum. Their chemical structures were elucidated using spectroscopic methods (UV, IR, ESI-Q-TOF-MS, and 1D/2D NMR). All the isolates were evaluated for their ability to inhibit IL-6 activity in the human embryonic kidney-Blue™ IL-6 cell line and their cytotoxic activity against ovarian cancer cell lines (SKOV3/SKOV3-TR) and chemotherapy-resistant variants (cisplatin-resistant A2780/paclitaxel-resistant SKOV3). The compounds 3, 4, 11, 12, 17, and 18 exhibited IL-6 inhibition comparable to that of the positive control bazedoxifene. Notably, compound 12 displayed the most potent anticancer effect against all the tested cancer cell lines. These findings highlight the importance of researching the diverse activities of both known and newly discovered natural products to fully unlock their potential therapeutic benefits.
Collapse
Affiliation(s)
- Hye Jin Kim
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (H.J.K.); (A.K.); (K.J.S.)
| | - Lee Kyung Kim
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, BK21FOUR Team for Advanced Program for Smart Pharma Leaders, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (L.K.K.); (T.-H.H.)
| | - Anna Kim
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (H.J.K.); (A.K.); (K.J.S.)
| | - Khin Myo Htwe
- Popa Mountain National Park, Forest Department, Kyaukpadaung Township, Mandalay Division, Kyaukpadaung 05241, Myanmar;
| | - Tae-Hwe Heo
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, BK21FOUR Team for Advanced Program for Smart Pharma Leaders, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (L.K.K.); (T.-H.H.)
| | - Kye Jung Shin
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (H.J.K.); (A.K.); (K.J.S.)
| | - Hee Jung Kim
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, BK21FOUR Team for Advanced Program for Smart Pharma Leaders, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (L.K.K.); (T.-H.H.)
| | - Kee Dong Yoon
- College of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (H.J.K.); (A.K.); (K.J.S.)
| |
Collapse
|
11
|
Nie D, Ma H, Huang G, Zhao T, Li W. Knowledge, Attitudes, and Practices of Physicians Regarding Targeted Drug Therapy for Lung Cancer. Int J Gen Med 2024; 17:2681-2689. [PMID: 38883701 PMCID: PMC11178084 DOI: 10.2147/ijgm.s465079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose This study aimed to examine the KAP of physicians regarding targeted drug therapy for lung cancer in China. Methods This cross-sectional study enrolled physicians working in hospitals in Nanyang. A self-administered questionnaire was developed (Cronbach's α=0.912) to collect the demographic information and KAP. Results This study included 191 valid questionnaires. Most participants were male (70.2%) and aged 36-50 (55.5%). The median knowledge score was 29 (24-31) (/36, 80.6%), the mean attitude score was 42 (39-44) (/50, 84.0%), and the mean practice score was 28 (26-29) (/30, 93.3%), indicating sufficient knowledge, positive attitudes, and proactive practice. The female gender (OR=5.291, 95% CI: 1.426-19.634, P=0.013), working in non-public tertiary hospitals (OR=0.053, 95% CI: 0.008-0.360, P=0.003), and working in medical oncology (OR=10.764, 95% CI: 2.638-43.922, P=0.001) were independently associated with adequate knowledge. Only the knowledge scores (OR=1.121, 95% CI: 1.036-1.212, P=0.004) were independently associated with a positive attitude. Only the attitude scores (OR=1.895, 95% CI: 1.333-2.694, P<0.001) were independently associated with proactive practice. Conclusion Physicians working in thoracic surgery, respiratory medicine, or medical oncology displayed sufficient knowledge, positive attitude, and proactive practice toward targeted therapy for lung cancer.
Collapse
Affiliation(s)
- Di Nie
- Thoracic Surgery Department, The First Affiliated Hospital of Nanyang Medical College, Nanyang, People's Republic of China
- Graduate Division Department, Xinxiang Medical University, Xinxiang, Chian
| | - Haozhi Ma
- Thoracic Surgery Department, The First Affiliated Hospital of Nanyang Medical College, Nanyang, People's Republic of China
- Graduate Division Department, Xinxiang Medical University, Xinxiang, Chian
| | - Guosheng Huang
- Thoracic Surgery Department, The First Affiliated Hospital of Nanyang Medical College, Nanyang, People's Republic of China
| | - Tianzeng Zhao
- Thoracic Surgery Department, The First Affiliated Hospital of Nanyang Medical College, Nanyang, People's Republic of China
| | - Wenxian Li
- Thoracic Surgery Department, The First Affiliated Hospital of Nanyang Medical College, Nanyang, People's Republic of China
| |
Collapse
|
12
|
Soares C, Abreu G, Nogueira da Silva TL, Queiroz J, Menezes P, Bernardino G, Pires T, Carrizo M, Felice R, Riggi MC, Cravero F, Ribeiro de Souza ALA, Jotimliansky L. OCEANIA: real-world study of ovarian cancer treatment patterns across multiple lines of therapy in Argentina and Brazil. Future Oncol 2024; 20:2023-2036. [PMID: 38861309 PMCID: PMC11526727 DOI: 10.1080/14796694.2024.2343650] [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: 09/11/2023] [Accepted: 04/12/2024] [Indexed: 06/12/2024] Open
Abstract
Aim: To evaluate real-world data on treatment patterns in Argentina and Brazil in patients with ovarian cancer.Methods: This study evaluated de-identified antineoplastic exposure data from a private healthcare provider in Argentina and health claims database (Orizon) in Brazil from 2010 to 2019 and 2015 to 2020, respectively.Results: Platinum-based chemotherapy was the most common first-line therapy (Argentina: n =311 [87.6%]; Brazil: n = 1142 [79.3%]). The proportion of patients receiving platinum-based chemotherapy declined across both populations from first- to second-line, while use of non-platinum-based, targeted, and hormone therapies increased. Duration of platinum-based treatment and time to next treatment decreased from first- to fourth-line.Conclusion: There is an unmet need for effective therapies that can prolong time to next treatment in ovarian cancer in Argentina and Brazil.
Collapse
|
13
|
Nunes M, Bartosch C, Abreu MH, Richardson A, Almeida R, Ricardo S. Deciphering the Molecular Mechanisms behind Drug Resistance in Ovarian Cancer to Unlock Efficient Treatment Options. Cells 2024; 13:786. [PMID: 38727322 PMCID: PMC11083313 DOI: 10.3390/cells13090786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Ovarian cancer is a highly lethal form of gynecological cancer. This disease often goes undetected until advanced stages, resulting in high morbidity and mortality rates. Unfortunately, many patients experience relapse and succumb to the disease due to the emergence of drug resistance that significantly limits the effectiveness of currently available oncological treatments. Here, we discuss the molecular mechanisms responsible for resistance to carboplatin, paclitaxel, polyadenosine diphosphate ribose polymerase inhibitors, and bevacizumab in ovarian cancer. We present a detailed analysis of the most extensively investigated resistance mechanisms, including drug inactivation, drug target alterations, enhanced drug efflux pumps, increased DNA damage repair capacity, and reduced drug absorption/accumulation. The in-depth understanding of the molecular mechanisms associated with drug resistance is crucial to unveil new biomarkers capable of predicting and monitoring the kinetics during disease progression and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Carla Bartosch
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (CI-IPO-Porto), Health Research Network (RISE@CI-IPO-Porto), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Miguel Henriques Abreu
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Alan Richardson
- The School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, Staffordshire, UK;
| | - Raquel Almeida
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Biology Department, Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
14
|
Mishra AK, Ye T, Banday S, Thakare RP, Su CTT, Pham NNH, Ali A, Kulshreshtha A, Chowdhury SR, Simone TM, Hu K, Zhu LJ, Eisenhaber B, Deibler SK, Simin K, Thompson PR, Kelliher MA, Eisenhaber F, Malonia SK, Green MR. Targeting the GPI transamidase subunit GPAA1 abrogates the CD24 immune checkpoint in ovarian cancer. Cell Rep 2024; 43:114041. [PMID: 38573857 DOI: 10.1016/j.celrep.2024.114041] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
CD24 is frequently overexpressed in ovarian cancer and promotes immune evasion by interacting with its receptor Siglec10, present on tumor-associated macrophages, providing a "don't eat me" signal that prevents targeting and phagocytosis by macrophages. Factors promoting CD24 expression could represent novel immunotherapeutic targets for ovarian cancer. Here, using a genome-wide CRISPR knockout screen, we identify GPAA1 (glycosylphosphatidylinositol anchor attachment 1), a factor that catalyzes the attachment of a glycosylphosphatidylinositol (GPI) lipid anchor to substrate proteins, as a positive regulator of CD24 cell surface expression. Genetic ablation of GPAA1 abolishes CD24 cell surface expression, enhances macrophage-mediated phagocytosis, and inhibits ovarian tumor growth in mice. GPAA1 shares structural similarities with aminopeptidases. Consequently, we show that bestatin, a clinically advanced aminopeptidase inhibitor, binds to GPAA1 and blocks GPI attachment, resulting in reduced CD24 cell surface expression, increased macrophage-mediated phagocytosis, and suppressed growth of ovarian tumors. Our study highlights the potential of targeting GPAA1 as an immunotherapeutic approach for CD24+ ovarian cancers.
Collapse
Affiliation(s)
- Alok K Mishra
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Tianyi Ye
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ritesh P Thakare
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chinh Tran-To Su
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore
| | - Ngoc N H Pham
- Faculty of Biology and Biotechnology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City, Vietnam
| | - Amjad Ali
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ankur Kulshreshtha
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shreya Roy Chowdhury
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tessa M Simone
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kai Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine and Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Birgit Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore; Lausitz Advanced Scientific Applications (LASA) gGmbH, Straße der Einheit 2-24, 02943 Weißwasser, Germany
| | - Sara K Deibler
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Karl Simin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michelle A Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore; Lausitz Advanced Scientific Applications (LASA) gGmbH, Straße der Einheit 2-24, 02943 Weißwasser, Germany; School of Biological Sciences, Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Sunil K Malonia
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Michael R Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
15
|
Lacy MS, Jenner AL. Impact of Resistance on Therapeutic Design: A Moran Model of Cancer Growth. Bull Math Biol 2024; 86:43. [PMID: 38502371 PMCID: PMC10950993 DOI: 10.1007/s11538-024-01272-6] [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: 09/04/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024]
Abstract
Resistance of cancers to treatments, such as chemotherapy, largely arise due to cell mutations. These mutations allow cells to resist apoptosis and inevitably lead to recurrence and often progression to more aggressive cancer forms. Sustained-low dose therapies are being considered as an alternative over maximum tolerated dose treatments, whereby a smaller drug dosage is given over a longer period of time. However, understanding the impact that the presence of treatment-resistant clones may have on these new treatment modalities is crucial to validating them as a therapeutic avenue. In this study, a Moran process is used to capture stochastic mutations arising in cancer cells, inferring treatment resistance. The model is used to predict the probability of cancer recurrence given varying treatment modalities. The simulations predict that sustained-low dose therapies would be virtually ineffective for a cancer with a non-negligible probability of developing a sub-clone with resistance tendencies. Furthermore, calibrating the model to in vivo measurements for breast cancer treatment with Herceptin, the model suggests that standard treatment regimens are ineffective in this mouse model. Using a simple Moran model, it is possible to explore the likelihood of treatment success given a non-negligible probability of treatment resistant mutations and suggest more robust therapeutic schedules.
Collapse
Affiliation(s)
- Mason S Lacy
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
16
|
Abujamous L, Soltani A, Al-Thawadi H, Agouni A. Advances in nanotechnology-enabled drug delivery for combining PARP inhibitors and immunotherapy in advanced ovarian cancer. BIOMOLECULES & BIOMEDICINE 2024; 24:230-237. [PMID: 38231530 PMCID: PMC10950340 DOI: 10.17305/bb.2023.9757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/25/2023] [Accepted: 11/23/2023] [Indexed: 01/18/2024]
Abstract
Advanced ovarian cancer is a malignancy that spreads beyond the ovaries to the pelvis, abdomen, lungs, or lymph nodes. Effective treatment options are available to improve survival rates in patients with advanced ovarian cancer. These include radiation, surgery, chemotherapy, immunotherapy, and targeted therapy. Drug resistance, however, remains a significant challenge in pharmacotherapeutic interventions, leading to reduced efficacy and unfavorable patient outcomes. Combination therapy, which involves using multiple drugs with different mechanisms of action at their optimal dose, is a promising approach to circumvent this challenge and it involves using multiple drugs with different mechanisms of action at their optimal dose. In recent years, nanotechnology has emerged as a valuable alternative for enhancing drug delivery precision and minimize toxicity. Nanoparticles can deliver drugs to specific cancer cells, resulting in higher drug concentrations at the tumor site, and reducing overall drug toxicity. Nanotechnology-based drug delivery systems have the potential to improve the therapeutic effects of anti-cancer drugs, reduce drug resistance, and improve outcomes for patients with advanced ovarian cancer. This literature review aims to examine the current understanding of combining poly (ADP-ribose) polymerase (PARP) inhibitors and immunotherapy in treating advanced ovarian cancer and the potential impact of nanotechnology on drug delivery.
Collapse
Affiliation(s)
- Lama Abujamous
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Office of Vice President for Research and Graduate Studies, Qatar University, Doha, Qatar
| | - Abderrezzaq Soltani
- Office of Vice President for Medical and Health Sciences, Qatar University, Doha, Qatar
- Department of Clinical Pharmacy and Practice, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Hamda Al-Thawadi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdelali Agouni
- Office of Vice President for Research and Graduate Studies, Qatar University, Doha, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
17
|
Tu YP, Hanze E, Zhu F, Lagraauw HM, Sloss CM, Method M, Esteves B, Westin EH, Berkenblit A. Population pharmacokinetics of mirvetuximab soravtansine in patients with folate receptor-α positive ovarian cancer: The antibody-drug conjugate, payload and metabolite. Br J Clin Pharmacol 2024; 90:568-581. [PMID: 37872122 DOI: 10.1111/bcp.15937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
AIMS Mirvetuximab soravtansine is a first-in-class antibody-drug conjugate recently approved for the treatment of folate receptor-α positive ovarian cancer. The aim of this study was to develop a population pharmacokinetic model to describe the concentration-time profiles of mirvetuximab soravtansine, the payload (DM4) and a metabolite (S-methyl-DM4). METHODS Mirvetuximab soravtansine was administered intravenously from 0.15 to 7 mg/kg to 543 patients with predominantly platinum-resistant ovarian cancer in 3 clinical studies, and the plasma drug concentrations were analysed using a nonlinear mixed-effects modelling approach. Stepwise covariate modelling was performed to identify covariates. RESULTS We developed a semi-mechanistic population pharmacokinetic model that included linear and nonlinear routes for the elimination of mirvetuximab soravtansine and a target compartment for the formation and disposition of the payload and metabolite in tumour cells. The clearance and volume of the central compartment were 0.0153 L/h and 2.63 L for mirvetuximab soravtansine, 8.83 L/h and 3.67 L for DM4, and 2.04 L/h and 6.3 L for S-methyl-DM4, respectively. Body weight, serum albumin and age were identified as statistically significant covariates. Exposures in patients with renal or hepatic impairment and who used concomitant cytochrome P450 (CYP) 3A4 inhibitors were estimated. CONCLUSION There is no need for dose adjustment due to covariate effects for mirvetuximab soravtansine administered at the recommended dose of 6 mg/kg based on adjusted ideal body weight. Dose adjustment is not required for patients with mild or moderate renal impairment, mild hepatic impairment, or when concomitant weak and moderate CYP3A4 inhibitors are used.
Collapse
Affiliation(s)
- Ya-Ping Tu
- Clinical Pharmacology, ImmunoGen, Waltham, Massachusetts, USA
| | | | - Fengying Zhu
- Bioanalysis, ImmunoGen, Waltham, Massachusetts, USA
| | | | - Callum M Sloss
- Translational Science, ImmunoGen, Waltham, Massachusetts, USA
| | - Michael Method
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Brooke Esteves
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Eric H Westin
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| | - Anna Berkenblit
- Clinical Development, ImmunoGen, Waltham, Massachusetts, USA
| |
Collapse
|
18
|
Padbury EH, Bálint Š, Carollo E, Carter DRF, Becker EBE. TRPC3 signalling contributes to the biogenesis of extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e132. [PMID: 38938673 PMCID: PMC11080740 DOI: 10.1002/jex2.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/29/2023] [Accepted: 12/08/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) contribute to a wide range of pathological processes including cancer progression, yet the molecular mechanisms underlying their biogenesis remain incompletely characterized. The development of tetraspanin-based pHluorin reporters has enabled the real-time analysis of EV release at the plasma membrane. Here, we employed CD81-pHluorin to investigate mechanisms of EV release in ovarian cancer (OC) cells and report a novel role for the Ca2+-permeable transient receptor potential (TRP) channel TRPC3 in EV-mediated communication. We found that specific activation of TRPC3 increased Ca2+ signalling in SKOV3 cells and stimulated an immediate increase in EV release. Ca2+-stimulants histamine and ionomycin likewise induced EV release, and imaging analysis revealed distinct stimulation-dependent temporal and spatial release dynamics. Interestingly, inhibition of TRPC3 attenuated histamine-stimulated Ca2+-entry and EV release, indicating that TRPC3 is likely to act downstream of histamine signalling in EV biogenesis. Furthermore, we found that direct activation of TRPC3 as well as the application of EVs derived from TRPC3-activated cells increased SKOV3 proliferation. Our data provides insights into the molecular mechanisms and dynamics underlying EV release in OC cells, proposing a key role for TRPC3 in EV biogenesis.
Collapse
Affiliation(s)
- Elise H. Padbury
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Štefan Bálint
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Emanuela Carollo
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
| | - David R. F. Carter
- Department of Biological and Medical SciencesOxford Brookes UniversityOxfordUK
- Evox Therapeutics LimitedOxfordUK
| | - Esther B. E. Becker
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| |
Collapse
|
19
|
Gasimli K, Raab M, Mandal R, Krämer A, Peña-Llopis S, Tahmasbi Rad M, Becker S, Strebhardt K, Sanhaji M. Synergistic Sensitization of High-Grade Serous Ovarian Cancer Cells Lacking Caspase-8 Expression to Chemotherapeutics Using Combinations of Small-Molecule BRD4 and CDK9 Inhibitors. Cancers (Basel) 2023; 16:107. [PMID: 38201534 PMCID: PMC10778249 DOI: 10.3390/cancers16010107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers worldwide, with approximately 70% of cases diagnosed in advanced stages. This late diagnosis results from the absence of early warning symptoms and is associated with an unfavorable prognosis. A standard treatment entails a combination of primary chemotherapy with platinum and taxane agents. Tumor recurrence following first-line chemotherapy with Carboplatin and Paclitaxel is detected in 80% of advanced ovarian cancer patients, with disease relapse occurring within 2 years of initial treatment. Platinum-resistant ovarian cancer is one of the biggest challenges in treating patients. Second-line treatments involve PARP or VEGF inhibitors. Identifying novel biomarkers and resistance mechanisms is critical to overcoming resistance, developing newer treatment strategies, and improving patient survival. In this study, we have determined that low Caspase-8 expression in ovarian cancer patients leads to poor prognosis. High-Grade Serous Ovarian Cancer (HGSOC) cells lacking Caspase-8 expression showed an altered composition of the RNA Polymerase II-containing transcriptional elongation complex leading to increased transcriptional activity. Caspase-8 knockout cells display increased BRD4 expression and CDK9 activity and reduced sensitivities to Carboplatin and Paclitaxel. Based on our work, we are proposing three potential therapeutic approaches to treat advanced ovarian cancer patients who exhibit low Caspase-8 expression and resistance to Carboplatin and/or Paclitaxel-combinations of (1) Carboplatin with small-molecule BRD4 inhibitors; (2) Paclitaxel with small-molecule BRD4 inhibitors, and (3) small-molecule BRD4 and CDK9 inhibitors. In addition, we are also proposing two predictive markers of chemoresistance-BRD4 and pCDK9.
Collapse
Affiliation(s)
- Khayal Gasimli
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Monika Raab
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Ranadip Mandal
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Andrea Krämer
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Samuel Peña-Llopis
- Translational Genomics, Department of Ophthalmology, University Hospital Essen, 45147 Essen, Germany;
- German Cancer Consortium (DKTK), 45147 Essen, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Morva Tahmasbi Rad
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Sven Becker
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| | - Klaus Strebhardt
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
| | - Mourad Sanhaji
- Department of Gynecology, University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany; (K.G.); (M.R.); (R.M.); (A.K.)
| |
Collapse
|
20
|
Zhang Z, Zhang Q, Yu Y, Su S. Epigallocatechin gallate inhibits ovarian cancer cell growth and induces cell apoptosis via activation of FOXO3A. In Vitro Cell Dev Biol Anim 2023; 59:739-746. [PMID: 38038884 DOI: 10.1007/s11626-023-00830-x] [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: 04/12/2023] [Accepted: 08/12/2023] [Indexed: 12/02/2023]
Abstract
Epigallocatechin gallate (EGCG), a bioactive component in tea, displays broad anti-cancer effects. Our study was designed to evaluate the anti-cancer effects of EGCG on ovarian cancer and explored the underlying molecular mechanisms. To evaluate the in vitro inhibitory effects of EGCG against ovarian cancer, MTT assay, colony formation assay, apoptosis assay, and wound healing assay, were performed. Besides, the inhibitory effects of EGCG on tumor growth in the xenograft animal model were evaluated by measuring tumor volume and tumor weight. Moreover, Western blotting and qPCR were used to evaluate the levels of target genes and proteins. Treatment with EGCG inhibited cell migration and cell survival, and promoted cell apoptosis in A2780 and SKOV3 cells. Interestingly, treatment with EGCG inhibited the tumor growth in the xenograft animal model. The mechanistic study revealed that treatment with EGCG induced the activation of FOXO3A and suppressed the expression of c-Myc both in vitro and in vivo. Our findings demonstrate that EGCG suppress ovarian cancer cell growth, which may be due to its regulation on FOXO3A and c-Myc.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Qinghua Zhang
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Yani Yu
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Shan Su
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China.
| |
Collapse
|
21
|
Alarcon-Zapata P, Perez AJ, Toledo-Oñate K, Contreras H, Ormazabal V, Nova-Lamperti E, Aguayo CA, Salomon C, Zuniga FA. Metabolomics profiling and chemoresistance mechanisms in ovarian cancer cell lines: Implications for targeting glutathione pathway. Life Sci 2023; 333:122166. [PMID: 37827232 DOI: 10.1016/j.lfs.2023.122166] [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: 08/15/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
Ovarian cancer presents a significant challenge due to its high rate of chemoresistance, which complicates the effectiveness of drug-response therapy. This study provides a comprehensive metabolomic analysis of ovarian cancer cell lines OVCAR-3 and SK-OV-3, characterizing their distinct metabolic landscapes. Metabolomics coupled with chemometric analysis enabled us to discriminate between the metabolic profiles of these two cell lines. The OVCAR-3 cells, which are sensitive to doxorubicin (DOX), exhibited a preference for biosynthetic pathways associated with cell proliferation. Conversely, DOX-resistant SK-OV-3 cells favored fatty acid oxidation for energy maintenance. Notably, a marked difference in glutathione (GSH) metabolism was observed between these cell lines. Our investigations further revealed that GSH depletion led to a profound change in drug sensitivity, inducing a shift from a cytostatic to a cytotoxic response. The results derived from this comprehensive metabolomic analysis offer potential targets for novel therapeutic strategies to overcome drug resistance. Our study suggests that targeting the GSH pathway could potentially enhance chemotherapy's efficacy in treating ovarian cancer.
Collapse
Affiliation(s)
- Pedro Alarcon-Zapata
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile; Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Concepción, Chile
| | - Andy J Perez
- Department of Instrumental Analysis, Faculty of Pharmacy, University of Concepcion, Chile
| | - Karin Toledo-Oñate
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Hector Contreras
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Valeska Ormazabal
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepcion, Chile
| | - Estefania Nova-Lamperti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Claudio A Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane QLD 4029, Australia
| | - Felipe A Zuniga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Chile.
| |
Collapse
|
22
|
Huldani H, Abdul-Jabbar Ali S, Al-Dolaimy F, Hjazi A, Denis Andreevich N, Oudaha KH, Almulla AF, Alsaalamy A, Kareem Oudah S, Mustafa YF. The potential role of interleukins and interferons in ovarian cancer. Cytokine 2023; 171:156379. [PMID: 37757536 DOI: 10.1016/j.cyto.2023.156379] [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: 09/03/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
Ovarian cancer poses significant challenges and remains a highly lethal disease with limited treatment options. In the context of ovarian cancer, interleukins (ILs) and interferons (IFNs), important cytokines that play crucial roles in regulating the immune system, have emerged as significant factors influencing its development. This article provides a comprehensive review of the involvement of various ILs, including those from the IL-1 family, IL-2 family, IL-6 family, IL-8 family, IL-10 family, and IL-17 family, in ovarian cancer. The focus is on their impact on tumor growth, metastasis, and their role in evading immune responses within the tumor microenvironment. Additionally, the article conducts an in-depth examination of the oncogenic or antitumor roles of each IL in the context of ovarian cancer pathogenesis and progression. Besides, we elucidated the enhancements in the treatment of ovarian cancer through the utilization of type-I IFN and type-II IFN. Recent research has shed light on the intricate mechanisms through which specific ILs and IFNs contribute to the advancement of the disease. By incorporating recent findings, this review also seeks to inspire further investigations into unexplored mechanisms, fostering ongoing research to develop more effective therapeutic strategies for ovarian cancer. Moreover, through an in-depth analysis of IL- and IFN-associated clinical trials, we have highlighted their promising potential of in the treatment of ovarian cancer. These clinical trials serve to reinforce the significant outlook for utilizing ILs and IFNs as therapeutic agents in combating this disease.
Collapse
Affiliation(s)
- Huldani Huldani
- Department of Physiology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Khulood H Oudaha
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Abbas F Almulla
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
23
|
Szénási A, Sivasudhan E, Du H, Zhang P, Huang J, Zhang Z, Rocha S, Wang M. Targeting SOD1 via RNAi with PEGylated graphene oxide nanoparticles in platinum-resistant ovarian cancer. Cancer Gene Ther 2023; 30:1554-1568. [PMID: 37582934 PMCID: PMC10645591 DOI: 10.1038/s41417-023-00659-2] [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: 02/16/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Acquired platinum resistance poses a significant therapeutic impediment to ovarian cancer patient care, accounting for more than 200,000 deaths annually worldwide. We previously identified that overexpression of the antioxidant superoxide dismutase 1 (SOD1) in ovarian cancer is associated with a platinum-resistant phenotype via conferring oxidative stress resistance against platinum compounds. We further demonstrated that enzymatic inhibition using small-molecule inhibitors or silencing of SOD1 via RNA interference (RNAi) increased cisplatin sensitivity and potency in vitro. We launched this study to explore the potential therapeutic applications of SOD1 silencing in vivo in order to reverse cisplatin resistance using a graphene-based siRNA delivery platform. PEGylated graphene oxide (GO) polyethyleneimine (GOPEI-mPEG) nanoparticle was complexed with SOD1 siRNA. GOPEI-mPEG-siSOD1 exhibited high biocompatibility, siRNA loading capacity, and serum stability, and showed potent downregulation of SOD1 mRNA and protein levels. We further observed that cisplatin and PEI elicited mitochondrial dysfunction and transcriptionally activated the mitochondrial unfolded protein response (UPRmt) used as a reporter for their respective cytotoxicities. SOD1 silencing was found to augment cisplatin-induced cytotoxicity resulting in considerable tumour growth inhibition in cisplatin-sensitive A2780 and cisplatin-resistant A2780DDP subcutaneous mouse xenografts. Our study highlights the potential therapeutic applicability of RNAi-mediated targeting of SOD1 as a chemosensitizer for platinum-resistant ovarian cancers.
Collapse
Affiliation(s)
- Attila Szénási
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Enakshi Sivasudhan
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Hong Du
- Suzhou GenePharma, Suzhou, Jiangsu, 215123, China
| | | | - Jie Huang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Zhijun Zhang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Sonia Rocha
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Mu Wang
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China.
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
24
|
Nagumo Y, Villareal MO, Isoda H, Usui T. RSK4 confers paclitaxel resistance to ovarian cancer cells, which is resensitized by its inhibitor BI-D1870. Biochem Biophys Res Commun 2023; 679:23-30. [PMID: 37660640 DOI: 10.1016/j.bbrc.2023.08.060] [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: 07/03/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Many ovarian cancers initially respond well to chemotherapy, but often become drug-resistant after several years. Therefore, analysis of drug resistance mechanisms and overcoming resistance are urgently needed. Paclitaxel is one of the first-choice and widely-used drugs for ovarian cancer, but like most drugs, drug resistance is observed in subsequent use. RSK4 is known as a tumor-suppressor, however, it has increasingly been reported to lead to drug resistance. Here, we found that RSK4 expression was elevated in paclitaxel-resistant ovarian cancer cells using DNA microarray, quantitative real-time PCR, and western blotting analysis. We examined the contribution of RSK4 to paclitaxel resistance and found that paclitaxel sensitivity was restored by RSK inhibitor co-treatment. We analyzed the mechanism by which resistance is developed when RSK4 level is elevated, and accelerated phosphorylation of the downstream translation factor eIF4B was discovered. In the Kaplan-Meier plot, the overall survival time was longer with RSK4 high, supporting its role as a tumor suppressor, as in previous findings, but the tendency was reversed when focusing on paclitaxel treatment. In addition, RSK4 levels were higher in non-responders than in responders in the ROC plotter. Finally, external expression of RSK4 in ovarian cancer cells increased the cell viability under paclitaxel treatment. These findings suggest that RSK4 may contribute to paclitaxel resistance, and that co-treatment with RSK4 inhibitors is effective treatment of paclitaxel-resistant ovarian cancer in which RSK4 is elevated.
Collapse
Affiliation(s)
- Yoko Nagumo
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan; Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, 305-8572, Japan.
| | - Myra O Villareal
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
| | - Hiroko Isoda
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan; Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Takeo Usui
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan; Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
25
|
Pascual-Antón L, Sandoval P, González-Mateo GT, Kopytina V, Tomero-Sanz H, Arriero-País EM, Jiménez-Heffernan JA, Fabre M, Egaña I, Ferrer C, Simón L, González-Cortijo L, Sainz de la Cuesta R, López-Cabrera M. Targeting carcinoma-associated mesothelial cells with antibody-drug conjugates in ovarian carcinomatosis. J Pathol 2023; 261:238-251. [PMID: 37555348 DOI: 10.1002/path.6170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023]
Abstract
Ovarian carcinomatosis is characterized by the accumulation of carcinoma-associated mesothelial cells (CAMs) in the peritoneal stroma and mainly originates through a mesothelial-to-mesenchymal transition (MMT) process. MMT has been proposed as a therapeutic target for peritoneal metastasis. Most ovarian cancer (OC) patients present at diagnosis with peritoneal seeding, which makes tumor progression control difficult by MMT modulation. An alternative approach is to use antibody-drug conjugates (ADCs) targeted directly to attack CAMs. This strategy could represent the cornerstone of precision-based medicine for peritoneal carcinomatosis. Here, we performed complete transcriptome analyses of ascitic fluid-isolated CAMs in advanced OC patients with primary-, high-, and low-grade, serous subtypes and following neoadjuvant chemotherapy. Our findings suggest that both cancer biological aggressiveness and chemotherapy-induced tumor mass reduction reflect the MMT-associated changes that take place in the tumor surrounding microenvironment. Accordingly, MMT-related genes, including fibroblast activation protein (FAP), mannose receptor C type 2 (MRC2), interleukin-11 receptor alpha (IL11RA), myristoylated alanine-rich C-kinase substrate (MARCKS), and sulfatase-1 (SULF1), were identified as specific actionable targets in CAMs of OC patients, which is a crucial step in the de novo design of ADCs. These cell surface target receptors were also validated in peritoneal CAMs of colorectal cancer peritoneal implants, indicating that ADC-based treatment could extend to other abdominal tumors that show peritoneal colonization. As proof of concept, a FAP-targeted ADC reduced tumor growth in an OC xenograft mouse model with peritoneal metastasis-associated fibroblasts. In summary, we propose MMT as a potential source of ADC-based therapeutic targets for peritoneal carcinomatosis. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lucía Pascual-Antón
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Pilar Sandoval
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Guadalupe T González-Mateo
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Valeria Kopytina
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Henar Tomero-Sanz
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Eva María Arriero-País
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | | | | | | | | | | | | | | | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| |
Collapse
|
26
|
Macieja A, Gulbas I, Popławski T. DNA Double-Strand Break Repair Inhibitors: YU238259, A12B4C3 and DDRI-18 Overcome the Cisplatin Resistance in Human Ovarian Cancer Cells, but Not under Hypoxia Conditions. Curr Issues Mol Biol 2023; 45:7915-7932. [PMID: 37886943 PMCID: PMC10605129 DOI: 10.3390/cimb45100500] [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: 09/14/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Cisplatin (CDDP) is the cornerstone of standard treatment for ovarian cancer. However, the resistance of ovarian cancer cells to CDDP leads to an inevitable recurrence. One of the strategies to overcome resistance to CDDP is the combined treatment of ovarian cancer with CDDP and etoposide (VP-16), although this strategy is not always effective. This article presents a new approach to sensitize CDDP-resistant human ovarian carcinoma cells to combined treatment with CDDP and VP-16. To replicate the tumor conditions of cancers, we performed analysis under hypoxia conditions. Since CDDP and VP-16 induce DNA double-strand breaks (DSB), we introduce DSB repair inhibitors to the treatment scheme. We used novel HRR and NHEJ inhibitors: YU238259 inhibits the HRR pathway, and DDRI-18 and A12B4C3 act as NHEJ inhibitors. All inhibitors enhanced the therapeutic effect of the CDDP/VP-16 treatment scheme and allowed a decrease in the effective dose of CDDP/VP16. Inhibition of HRR or NHEJ decreased survival and increased DNA damage level, increased the amount of γ-H2AX foci, and caused an increase in apoptotic fraction after treatment with CDDP/VP16. Furthermore, delayed repair of DSBs was detected in HRR- or NHEJ-inhibited cells. This favorable outcome was altered under hypoxia, during which alternation at the transcriptome level of the transcriptome in cells cultured under hypoxia compared to aerobic conditions. These changes suggest that it is likely that other than classical DSB repair systems are activated in cancer cells during hypoxia. Our study suggests that the introduction of DSB inhibitors may improve the effectiveness of commonly used ovarian cancer treatment, and HRR, as well as NHEJ, is an attractive therapeutic target for overcoming the resistance to CDDP resistance of ovarian cancer cells. However, a hypoxia-mediated decrease in response to our scheme of treatment was observed.
Collapse
Affiliation(s)
- Anna Macieja
- Department of Microbiology and Pharmaceutical Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland;
| | - Izabela Gulbas
- Department of Immunology and Allergy, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland;
| | - Tomasz Popławski
- Department of Microbiology and Pharmaceutical Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland;
| |
Collapse
|
27
|
Drumond-Bock AL, Wang L, Wang L, Cybula M, Rostworowska M, Kinter M, Bieniasz M. Increased expression of BRD4 isoforms long (BRD4-L) and short (BRD4-S) promotes chemotherapy resistance in high-grade serous ovarian carcinoma. Genes Cancer 2023; 14:56-76. [PMID: 37705995 PMCID: PMC10496930 DOI: 10.18632/genesandcancer.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
Chemoresistance in ovarian carcinoma is a puzzling issue that urges understanding of strategies used by cancer cells to survive DNA damage and to escape cell death. Expanding efforts to understand mechanisms driving chemoresistance and to develop alternative therapies targeting chemoresistant tumors are critical. Amplification of BRD4 is frequently associated with chemoresistant ovarian carcinoma, but little is known about the biological effects of the overexpression of BRD4 isoforms in this malignancy. Here, we described the consequences of BRD4-L and BRD4-S overexpression in ovarian carcinoma shedding a light on a complex regulation of BRD4 isoforms. We demonstrated that the BRD4-L transcript expression is required to generate both isoforms, BRD4-L and BRD4-S. We showed that the BRD4-S mRNA expression positively correlated with BRD4-S protein levels, while BRD4-L isoform showed negative correlation between mRNA and protein levels. Moreover, we demonstrated that an overexpression of BRD4 isoforms is associated with chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Ana Luiza Drumond-Bock
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Luyao Wang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Lin Wang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | - Maria Rostworowska
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Magdalena Bieniasz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
28
|
Jazwinska DE, Kulawiec DG, Zervantonakis IK. Cancer-mesothelial and cancer-macrophage interactions in the ovarian cancer microenvironment. Am J Physiol Cell Physiol 2023; 325:C721-C730. [PMID: 37545408 PMCID: PMC10635648 DOI: 10.1152/ajpcell.00461.2022] [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: 10/12/2022] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
The metastatic ovarian cancer microenvironment is characterized by an intricate interaction network between cancer cells and host cells. This complex heterotypic cancer-host cell crosstalk results in an environment that promotes cancer cell metastasis and treatment resistance, leading to poor patient prognosis and survival. In this review, we focus on two host cell types found in the ovarian cancer microenvironment: mesothelial cells and tumor-associated macrophages. Mesothelial cells make up the protective lining of organs in the abdominal cavity. Cancer cells attach and invade through the mesothelial monolayer to form metastatic lesions. Crosstalk between mesothelial and cancer cells can contribute to metastatic progression and chemotherapy resistance. Tumor-associated macrophages are the most abundant immune cell type in the ovarian cancer microenvironment with heterogeneous subpopulations exhibiting protumor or antitumor functions. Macrophage reprogramming toward a protumor or antitumor state can be influenced by chemotherapy and communication with cancer cells, resulting in cancer cell invasion and treatment resistance. A better understanding of cancer-mesothelial and cancer-macrophage crosstalk will uncover biomarkers of metastatic progression and therapeutic targets to restore chemotherapy sensitivity.
Collapse
Affiliation(s)
- Dorota E Jazwinska
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Diana G Kulawiec
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Ioannis K Zervantonakis
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
29
|
Park J, Kim YS, Zhang S, Kim D, Shin S, Lee SH, Chung YJ. Single-cell RNA sequencing reveals a pro-metastatic subpopulation and the driver transcription factor NFE2L1 in ovarian cancer cells. Genes Genomics 2023; 45:1107-1115. [PMID: 37405595 DOI: 10.1007/s13258-023-01418-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023]
Abstract
BACKGROUND Although cytoreductive surgery followed by adjuvant chemotherapy is effective as a standard treatment for early-stage ovarian cancer, the majority of ovarian cancer cases are diagnosed at the advanced stages with dissemination to the peritoneal cavity, leading to a poor prognosis. Therefore, it is crucial to understand the cellular and molecular mechanisms underlying metastasis and identify novel therapeutic targets. OBJECTIVE In this study, we aimed to elucidate the mechanisms underlying gene expression alterations during the acquisition of metastatic potential and characterize the metastatic subpopulations within ovarian cancer cells. METHODS We conducted single-cell RNA sequencing of two human ovarian cancer cell lines: SKOV-3 and SKOV-3-13, a highly metastatic subclone of SKOV-3. Suppression of NFE2L1 expression was performed through siRNA-mediated knockdown and CRISPR-Cas9-mediated knockout. RESULTS Clustering and pseudotime trajectory analysis revealed pro-metastatic subpopulation within these cells. Furthermore, gene set enrichment analysis and prognosis analysis indicated that NFE2L1 could be a key transcription factor in the acquisition of metastasis potential. Inhibition of NFE2L1 significantly reduced migration and viability of both cells. In addition, NFE2L1 knockout cells exhibited significantly reduced tumor growth in a mouse xenograft model, recapitulating in silico and in vitro results. CONCLUSION The results presented in this study deepen our understanding of the molecular pathogenesis of ovarian cancer metastasis with the ultimate goal of developing treatments targeting pro-metastatic subclones prior to metastasis.
Collapse
Affiliation(s)
- Junseong Park
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Seob Kim
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Songzi Zhang
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Dokyeong Kim
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun Shin
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Sug Hyung Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Microbiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Qiu J, Li Z, An K, Niu L, Huang H, Xu F. Thermo-Chemical Resistance to Combination Therapy of Glioma Depends on Cellular Energy Level. ACS APPLIED MATERIALS & INTERFACES 2023; 15:39053-39063. [PMID: 37552210 DOI: 10.1021/acsami.3c05683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Thermal therapy has been widely used in clinical tumor treatment and more recently in combination with chemotherapy, where the key challenge is the treatment resistance. The mechanism at the cellular level underlying the resistance to thermo-chemical combination therapy remains elusive. In this study, we constructed 3D culture models for glioma cells (i.e., 3D glioma spheres) as the model system to recapitulate the native tumor microenvironment and systematically investigated the thermal response of 3D glioma spheres at different hyperthermic temperatures. We found that 3D glioma spheres show high viability under hyperthermia, especially under high hyperthermic temperatures (42 °C). Further study revealed that the main mechanism lies in the high energy level of cells in 3D glioma spheres under hyperthermia, which enables the cells to respond promptly to thermal stimulation and maintain cellular viability by upregulating the chaperon protein Hsp70 and the anti-apoptotic pathway AKT. Besides, we also demonstrated that 3D glioma spheres show strong drug resistance to the thermo-chemical combination therapy. This study provides a new perspective on understanding the thermal response of combination therapy for tumor treatment.
Collapse
Affiliation(s)
- Jinbin Qiu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhijie Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Keli An
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lele Niu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Haishui Huang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
31
|
Ara MG, Motalleb G, Velasco B, Rahdar A, Taboada P. Antineoplastic effect of paclitaxel-loaded polymeric nanocapsules on malignant human ovarian carcinoma cells (SKOV-3). J Mol Liq 2023; 384:122190. [DOI: 10.1016/j.molliq.2023.122190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
|
32
|
Tanabe S, Saigan M, Yoshimoto A, Sugino S, Ichida K, Niiya K, Morishima S. A case of hyponatremia attributed to carboplatin-induced syndrome of inappropriate anti-diuretic hormone. Gynecol Oncol Rep 2023; 48:101229. [PMID: 37389135 PMCID: PMC10300067 DOI: 10.1016/j.gore.2023.101229] [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: 04/25/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 07/01/2023] Open
Abstract
•A patient who used carboplatin in the treatment of ovarian cancer developed SIADH.•She was able to continue treatment with regular salt intake.•This treatment may be an option for similar patients in the future.
Collapse
Affiliation(s)
- Shohei Tanabe
- Corresponding author at: Kobe City Medical Center, West Hospital, 3-5-15-1001 Morigo-cho, Nada-ku, Kobe-shi, Hyogo 657-0028, Japan.
| | | | | | | | | | | | | |
Collapse
|
33
|
Zheng X, Wang X, Cheng X, Liu Z, Yin Y, Li X, Huang Z, Wang Z, Guo W, Ginhoux F, Li Z, Zhang Z, Wang X. Single-cell analyses implicate ascites in remodeling the ecosystems of primary and metastatic tumors in ovarian cancer. NATURE CANCER 2023; 4:1138-1156. [PMID: 37488416 PMCID: PMC10447252 DOI: 10.1038/s43018-023-00599-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Ovarian cancer (OC) is an aggressive gynecological tumor usually diagnosed with widespread metastases and ascites. Here, we depicted a single-cell landscape of the OC ecosystem with five tumor-relevant sites, including omentum metastasis and malignant ascites. Our data reveal the potential roles of ascites-enriched memory T cells as a pool for tumor-infiltrating exhausted CD8+ T cells and T helper 1-like cells. Moreover, tumor-enriched macrophages exhibited a preference for monocyte-derived ontogeny, whereas macrophages in ascites were more of embryonic origin. Furthermore, we characterized MAIT and dendritic cells in malignant ascites, as well as two endothelial subsets in primary tumors as predictive biomarkers for platinum-based chemotherapy response. Taken together, our study provides a global view of the female malignant ascites ecosystem and offers valuable insights for its connection with tumor tissues and paves the way for potential markers of efficacy evaluation and therapy resistance in OC.
Collapse
Affiliation(s)
- Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinjing Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xi Cheng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhaoyuan Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoduan Li
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | - Ziliang Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Guo
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florent Ginhoux
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Ziyi Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zemin Zhang
- BIOPIC, and School of Life Sciences, Peking University, Beijing, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
34
|
Zhang S, Kim D, Park M, Yin JH, Park J, Chung YJ. Suppression of Metastatic Ovarian Cancer Cells by Bepridil, a Calcium Channel Blocker. Life (Basel) 2023; 13:1607. [PMID: 37511982 PMCID: PMC10381520 DOI: 10.3390/life13071607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Although surgery followed by platinum-based therapy is effective as a standard treatment in the early stages of ovarian cancer, the majority of cases are diagnosed at advanced stages, leading to poor prognosis. Thus, the identification of novel therapeutic drugs is needed. In this study, we assessed the effectiveness of bepridil-a calcium channel blocker-in ovarian cancer cells using two cell lines: SKOV-3, and SKOV-3-13 (a highly metastatic clone of SKOV-3). Treatment of these cell lines with bepridil significantly reduced cell viability, migration, and invasion. Notably, SKOV-3-13 was more sensitive to bepridil than SKOV-3. The TGF-β1-induced epithelial-mesenchymal transition (EMT)-like phenotype was reversed by treatment with bepridil in both cell lines. Consistently, expression levels of EMT-related markers, including vimentin, β-catenin, and Snail, were also substantially decreased by the treatment with bepridil. An in vivo mouse xenograft model was used to confirm these findings. Tumor growth was significantly reduced by bepridil treatment in SKOV-3-13-inoculated mice, and immunohistochemistry showed consistently decreased expression of EMT-related markers. Our findings are the first to report anticancer effects of bepridil in ovarian cancer, and they suggest that bepridil holds significant promise as an effective therapeutic agent for targeting metastatic ovarian cancer.
Collapse
Affiliation(s)
- Songzi Zhang
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dokyeong Kim
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Minyoung Park
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jing Hu Yin
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junseong Park
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yeun-Jun Chung
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
35
|
Kelly R, Aviles D, Krisulevicz C, Hunter K, Krill L, Warshal D, Ostrovsky O. The Effects of Natural Epigenetic Therapies in 3D Ovarian Cancer and Patient-Derived Tumor Explants: New Avenues in Regulating the Cancer Secretome. Biomolecules 2023; 13:1066. [PMID: 37509102 PMCID: PMC10377145 DOI: 10.3390/biom13071066] [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: 05/04/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
High mortality rates in ovarian cancer have been linked to recurrence, metastasis, and chemoresistant disease, which are known to involve not only genetic changes but also epigenetic aberrations. In ovarian cancer, adipose-derived stem cells from the omentum (O-ASCs) play a crucial role in supporting the tumor and its tumorigenic microenvironment, further propagating epigenetic abnormalities and dissemination of the disease. Epigallocatechin gallate (EGCG), a DNA methyltransferase inhibitor derived from green tea, and Indole-3-carbinol (I3C), a histone deacetylase inhibitor from cruciferous vegetables, carry promising effects in reprograming aberrant epigenetic modifications in cancer. Therefore, we demonstrate the action of these diet-derived compounds in suppressing the growth of 3D ovarian cancer spheroids or organoids as well as post-treatment cancer recovery through proliferation, migration, invasion, and colony formation assays when compared to the synthetic epigenetic compound Panobinostat with or without standard chemotherapy. Finally, given the regulatory role of the secretome in growth, metastasis, chemoresistance, and relapse of disease, we demonstrate that natural epigenetic compounds can regulate the secretion of protumorigenic growth factors, cytokines, extracellular matrix components, and immunoregulatory markers in human ovarian cancer specimens. While further studies are needed, our results suggest that these treatments could be considered in the future as adjuncts to standard chemotherapy, improving efficiency and patient outcomes.
Collapse
Affiliation(s)
- Rebeca Kelly
- Department of Gynecologic Oncology, MD Anderson Cancer Center at Cooper University Hospital, Camden, NJ 08103, USA
| | - Diego Aviles
- Department of Gynecologic Oncology, MD Anderson Cancer Center at Cooper University Hospital, Camden, NJ 08103, USA
| | | | - Krystal Hunter
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Cooper Research Institute, Cooper University Healthcare, Camden, NJ 08103, USA
| | - Lauren Krill
- Department of Gynecologic Oncology, MD Anderson Cancer Center at Cooper University Hospital, Camden, NJ 08103, USA
| | - David Warshal
- Department of Gynecologic Oncology, MD Anderson Cancer Center at Cooper University Hospital, Camden, NJ 08103, USA
| | - Olga Ostrovsky
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Cooper Research Institute, Cooper University Healthcare, Camden, NJ 08103, USA
| |
Collapse
|
36
|
Alfano A, Cafferata EGA, Gangemi M, Nicola Candia A, Malnero CM, Bermudez I, Lopez MV, Ríos GD, Rotondaro C, Cuneo N, Curiel DT, Podhajcer OL, Lopez MV. In Vitro and In Vivo Efficacy of a Stroma-Targeted, Tumor Microenvironment Responsive Oncolytic Adenovirus in Different Preclinical Models of Cancer. Int J Mol Sci 2023; 24:9992. [PMID: 37373140 PMCID: PMC10297998 DOI: 10.3390/ijms24129992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/27/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
More than one million women are diagnosed annually worldwide with a gynecological cancer. Most gynecological cancers are diagnosed at a late stage, either because a lack of symptoms, such as in ovarian cancer or limited accessibility to primary prevention in low-resource countries, such as in cervical cancer. Here, we extend the studies of AR2011, a stroma-targeted and tumor microenvironment responsive oncolytic adenovirus (OAdV), whose replication is driven by a triple hybrid promoter. We show that AR2011 was able to replicate and lyse in vitro fresh explants obtained from human ovarian cancer, uterine cancer, and cervical cancer. AR2011 was also able to strongly inhibit the in vitro growth of ovarian malignant cells obtained from human ascites fluid. The virus could synergize in vitro with cisplatin even on ascites-derived cells obtained from patients heavily pretreated with neoadjuvant chemotherapy. AR2011(h404), a dual transcriptionally targeted derived virus armed with hCD40L and h41BBL under the regulation of the hTERT promoter, showed a strong efficacy in vivo both on subcutaneous and intraperitoneally established human ovarian cancer in nude mice. Preliminary studies in an immunocompetent murine tumor model showed that AR2011(m404) expressing the murine cytokines was able to induce an abscopal effect. The present studies suggest that AR2011(h404) is a likely candidate as a novel medicine for intraperitoneal disseminated ovarian cancer.
Collapse
Affiliation(s)
- Ana Alfano
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Eduardo G. A. Cafferata
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Mariela Gangemi
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Alejandro Nicola Candia
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Cristian M. Malnero
- Facultad de Ingeniería, Universidad Argentina de la Empresa, Lima 775, Ciudad Autónoma de Buenos Aires C1073AAO, Argentina; (C.M.M.); (I.B.)
| | - Ismael Bermudez
- Facultad de Ingeniería, Universidad Argentina de la Empresa, Lima 775, Ciudad Autónoma de Buenos Aires C1073AAO, Argentina; (C.M.M.); (I.B.)
| | - Mauricio Vargas Lopez
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Gregorio David Ríos
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Cecilia Rotondaro
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Nicasio Cuneo
- Servicio de Ginecología, Departamento de Cirugía, Hospital Municipal de Oncología Maria Curie, Avenida Patricias Argentinas 750, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina;
| | - David T. Curiel
- Division of Cancer Biology, Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA;
| | - Osvaldo L. Podhajcer
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| | - Maria Veronica Lopez
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir, IIBBA-CONICET, Avenida Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina; (A.A.); (E.G.A.C.); (M.G.); (A.N.C.); (M.V.L.); (G.D.R.); (C.R.)
| |
Collapse
|
37
|
Parvini S, Majidpoor J, Mortezaee K. The impact of PD-L1 as a biomarker of cancer responses to combo anti-PD-1/CTLA-4. Pathol Res Pract 2023; 247:154583. [PMID: 37267723 DOI: 10.1016/j.prp.2023.154583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
Combination therapy of solid tumors with immune checkpoint inhibitors (ICIs) is a promising and rapidly evolving area of clinical research. Combo nivolumab-ipilimumab therapy has demonstrated potent efficacy in recent years, and PD-L1 expression profile has shown to play a key role in determining the most optimal immunotherapeutic regimen in advanced cancer patients. Here, the focus is over the impact of PD-L1 on combo nivolumab-ipilimumab in advanced solid cancer patients. Interpretations of this review indicate that patient responses to combo nivolumab-ipilimumab can be affected from different levels of PD-L1 expression states. A point required attention is the variations in responses among diverse cancer types or between different doses of the immunotherapy drugs. In general, higher rates of responses are seen with higher PD-L1 expression levels in many cancer types. This, however, is not coincided with survival of patients. Taken all into consideration, it could be asserted that considering PD-L1 as a solo biomarker may not be reliable for predicting clinical efficacy of combo nivolumab-ipilimumab. Thus, a search for other biomarkers or combination of PD-L1 with other factors may be considered for predicting patient responses.
Collapse
Affiliation(s)
- Sasan Parvini
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
38
|
Hankittichai P, Thaklaewphan P, Wikan N, Ruttanapattanakul J, Potikanond S, Smith DR, Nimlamool W. Resveratrol Enhances Cytotoxic Effects of Cisplatin by Inducing Cell Cycle Arrest and Apoptosis in Ovarian Adenocarcinoma SKOV-3 Cells through Activating the p38 MAPK and Suppressing AKT. Pharmaceuticals (Basel) 2023; 16:ph16050755. [PMID: 37242538 DOI: 10.3390/ph16050755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
In the current study, we identified a mechanism of resveratrol (RES) underlying its anti-cancer properties against human ovarian adenocarcinoma SKOV-3 cells. We investigated its anti-proliferative and apoptosis-inducing effects in combination with cisplatin, using cell viability assay, flow cytometry, immunofluorescence study and Western blot analysis. We discovered that RES suppressed cancer cell proliferation and stimulated apoptosis, especially when combined with cisplatin. This compound also inhibited SKOV-3 cell survival, which may partly be due to its potential to inhibit protein kinase B (AKT) phosphorylation and induce the S-phase cell cycle arrest. RES in combination with cisplatin strongly induced cancer cell apoptosis through activating the caspase-dependent cascade, which was associated with its ability to stimulate nuclear phosphorylation of p38 mitogen-activated protein kinase (MAPK), well recognized to be involved in transducing environmental stress signals. RES-induced p38 phosphorylation was very specific, and the activation status of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) was not mainly affected. Taken together, our study provides accumulated evidence that RES represses proliferation and promotes apoptosis in SKOV-3 ovarian cancer cells through activating the p38 MAPK pathway. It is interesting that this active compound may be used as an effective agent to sensitize ovarian cancer to apoptosis induced by standard chemotherapies.
Collapse
Affiliation(s)
- Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
39
|
Bhat AA, Thapa R, Afzal O, Agrawal N, Almalki WH, Kazmi I, Alzarea SI, Altamimi ASA, Prasher P, Singh SK, Dua K, Gupta G. The pyroptotic role of Caspase-3/GSDME signalling pathway among various cancer: A Review. Int J Biol Macromol 2023; 242:124832. [PMID: 37196719 DOI: 10.1016/j.ijbiomac.2023.124832] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Cytotoxic drugs have long been recognised to kill cancer cells through apoptosis. According to a current study, pyroptosis inhibits cell proliferation and shrinks tumors. Pyroptosis and apoptosis are caspase-dependent programmed cell death (PCD) processes. Inflammasomes activate caspase-1 and latent cytokines, including IL-1β and IL-18, to cleave gasdermin E (GSDME) and induce pyroptosis. Gasdermin proteins activate caspase-3 to induce pyroptosis, which is associated with tumour genesis, development, and therapy response. These proteins may serve as therapeutic biomarkers for cancer detection, and their antagonists may be a new target. Caspase-3, a crucial protein in both pyroptosis and apoptosis, governs tumour cytotoxicity when activated, and GSDME expression modulates this. Once active caspase-3 cleaves GSDME, its N-terminal domain punches holes in the cell membrane, causing it to expand, burst, and die. To understand the cellular and molecular mechanisms of PCD mediated by caspase-3 and GSDME, we focused on pyroptosis. Hence, caspase-3 and GSDME may be promising targets for cancer treatment.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, U. P., India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun 248007, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
40
|
Mathews J, Chang A(J, Devlin L, Levin M. Cellular signaling pathways as plastic, proto-cognitive systems: Implications for biomedicine. PATTERNS (NEW YORK, N.Y.) 2023; 4:100737. [PMID: 37223267 PMCID: PMC10201306 DOI: 10.1016/j.patter.2023.100737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Many aspects of health and disease are modeled using the abstraction of a "pathway"-a set of protein or other subcellular activities with specified functional linkages between them. This metaphor is a paradigmatic case of a deterministic, mechanistic framework that focuses biomedical intervention strategies on altering the members of this network or the up-/down-regulation links between them-rewiring the molecular hardware. However, protein pathways and transcriptional networks exhibit interesting and unexpected capabilities such as trainability (memory) and information processing in a context-sensitive manner. Specifically, they may be amenable to manipulation via their history of stimuli (equivalent to experiences in behavioral science). If true, this would enable a new class of biomedical interventions that target aspects of the dynamic physiological "software" implemented by pathways and gene-regulatory networks. Here, we briefly review clinical and laboratory data that show how high-level cognitive inputs and mechanistic pathway modulation interact to determine outcomes in vivo. Further, we propose an expanded view of pathways from the perspective of basal cognition and argue that a broader understanding of pathways and how they process contextual information across scales will catalyze progress in many areas of physiology and neurobiology. We argue that this fuller understanding of the functionality and tractability of pathways must go beyond a focus on the mechanistic details of protein and drug structure to encompass their physiological history as well as their embedding within higher levels of organization in the organism, with numerous implications for data science addressing health and disease. Exploiting tools and concepts from behavioral and cognitive sciences to explore a proto-cognitive metaphor for the pathways underlying health and disease is more than a philosophical stance on biochemical processes; at stake is a new roadmap for overcoming the limitations of today's pharmacological strategies and for inferring future therapeutic interventions for a wide range of disease states.
Collapse
Affiliation(s)
- Juanita Mathews
- Allen Discovery Center at Tufts University, Medford, MA, USA
| | | | - Liam Devlin
- Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| |
Collapse
|
41
|
Feely N, Wdowicz A, Chevalier A, Wang Y, Li P, Rollo F, Lee GU. Targeting Mucin Protein Enables Rapid and Efficient Ovarian Cancer Cell Capture: Role of Nanoparticle Properties in Efficient Capture and Culture. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207154. [PMID: 36772896 DOI: 10.1002/smll.202207154] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/18/2023] [Indexed: 05/04/2023]
Abstract
The development of specific and sensitive immunomagnetic cell separation nanotechnologies is central to enhancing the diagnostic relevance of circulating tumor cells (CTCs) and improving cancer patient outcomes. The limited number of specific biomarkers used to enrich a phenotypically diverse set of CTCs from liquid biopsies has limited CTC yields and purity. The ultra-high molecular weight mucin, mucin16 (MUC16) is shown to physically shield key membrane proteins responsible for activating immune responses against ovarian cancer cells and may interfere with the binding of magnetic nanoparticles to popular immunomagnetic cell capture antigens. MUC16 is expressed in ≈90% of ovarian cancers and is almost universal in High Grade Serous Epithelial Ovarian Cancer. This work demonstrates that cell bound MUC16 is an effective target for rapid immunomagnetic extraction of expressor cells with near quantitative yield, high purity and viability from serum. The results provide a mechanistic insight into the effects of nanoparticle physical properties and immunomagnetic labeling on the efficiency of immunomagnetic cell isolation. The growth of these cells has also been studied after separation, demonstrating that nanoparticle size impacts cell-particle behavior and growth rate. These results present the successful isolation of "masked" CTCs enabling new strategies for the detection of cancer recurrence and select and monitor chemotherapy.
Collapse
Affiliation(s)
- Nathan Feely
- Conway Institute for Biomedical Research and School of Chemistry, University College Dublin, 61 Adair, Sandymount Ave, Dublin, CO. DUBLIN, 00004, Ireland
| | - Anita Wdowicz
- Conway Institute for Biomedical Research and School of Chemistry, University College Dublin, 61 Adair, Sandymount Ave, Dublin, CO. DUBLIN, 00004, Ireland
| | - Anne Chevalier
- Magnostics Ltd, 2 Clifton Lane, Merrion Road, Monkstown, Dublin, A94 A306, Ireland
| | - Ying Wang
- Magnostics Ltd, 2 Clifton Lane, Merrion Road, Monkstown, Dublin, A94 A306, Ireland
| | - Peng Li
- Magnostics Ltd, 2 Clifton Lane, Merrion Road, Monkstown, Dublin, A94 A306, Ireland
| | - Fanny Rollo
- École nationale supérieure des ingénieurs en arts chimiques et technologiques, Toulouse, 31030, France
| | - Gil U Lee
- Conway Institute for Biomedical Research and School of Chemistry, University College Dublin, 61 Adair, Sandymount Ave, Dublin, CO. DUBLIN, 00004, Ireland
| |
Collapse
|
42
|
Bhuia MS, Wilairatana P, Chowdhury R, Rakib AI, Kamli H, Shaikh A, Coutinho HDM, Islam MT. Anticancer Potentials of the Lignan Magnolin: A Systematic Review. Molecules 2023; 28:3671. [PMID: 37175081 PMCID: PMC10180476 DOI: 10.3390/molecules28093671] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Magnolin is a naturally occurring, multi-bioactive lignan molecule with inherent anticancer effects. This study aims to summarize the botanical origins and anticancer properties of magnolin. For this, a recent (as of March 2023) literature review was conducted using various academic search engines, including PubMed, Springer Link, Wiley Online, Web of Science, Science Direct, and Google Scholar. All the currently available information about this phytochemical and its role in various cancer types has been gathered and investigated. Magnolin is a compound found in many different plants. It has been demonstrated to have anticancer activity in numerous experimental models by inhibiting the cell cycle (G1 and G2/M phase); inducing apoptosis; and causing antiinvasion, antimetastasis, and antiproliferative effects via the modulation of several pathways. In conclusion, magnolin showed robust anticancer activity against many cancer cell lines by altering several cancer signaling pathways in various non- and pre-clinical experimental models, making it a promising plant-derived chemotherapeutic option for further clinical research.
Collapse
Affiliation(s)
- Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.)
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.)
| | - Asraful Islam Rakib
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.)
| | - Hossam Kamli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmad Shaikh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Henrique D. M. Coutinho
- Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.)
| |
Collapse
|
43
|
Wang Q, Cheng Y, Wang W, Tang X, Yang Y. Polyetherimide- and folic acid-modified Fe 3 O 4 nanospheres for enhanced magnetic hyperthermia performance. J Biomed Mater Res B Appl Biomater 2023; 111:795-804. [PMID: 36382676 DOI: 10.1002/jbm.b.35190] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/20/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022]
Abstract
Recent studies have highlighted the development prospects of magnetic hyperthermia in cancer therapy. A few studies on the application of Fe3 O4 nanospheres for the magnetic hyperthermia of gynecological malignancies have achieved certain efficacy, but there was no visible progress currently. In this work, Fe3 O4 nanospheres modified with polyetherimide (PEI) and folic acid (FA) were synthesized using a hydrothermal method for possible utility in biocompatible and active tumor-targeting magnetic induction hyperthermia. The PEI- and FA-coated Fe3 O4 nanospheres showed high crystallinity, well-dispersed spherical structures and ideal Ms value. As a result, the designed Fe3 O4 @ PEI@FA nanospheres achieved higher specific absorption rate (SAR) values at 360 kHz and 308 Oe, as well as excellent biocompatibility in Hela, SKOV3, HEC-1-A and NIH3T3 cells. These nanospheres can be used as an optimal heating agent for the magnetic hyperthermia treatment of gynecological cancers.
Collapse
Affiliation(s)
- Qinganzi Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gynecological Oncology Gansu Province, Lanzhou, China
| | - Yuemei Cheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gynecological Oncology Gansu Province, Lanzhou, China
| | - Wenhua Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gynecological Oncology Gansu Province, Lanzhou, China
| | - Xiaolin Tang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Key Laboratory for Gynecological Oncology Gansu Province, Lanzhou, China.,The Third People's Hospital of Gansu Province, Lanzhou, China
| | - Yongxiu Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gynecological Oncology Gansu Province, Lanzhou, China
| |
Collapse
|
44
|
Asouli A, Sadr S, Mohebalian H, Borji H. Anti-Tumor Effect of Protoscolex Hydatid Cyst Somatic Antigen on Inhibition Cell Growth of K562. Acta Parasitol 2023:10.1007/s11686-023-00680-3. [PMID: 36991291 DOI: 10.1007/s11686-023-00680-3] [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: 10/31/2022] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Today, cancer is one of the most important causes of death in the world, and so far, many treatment methods have been used in this field. Immunotherapy is considered one of the newest developments in this science, and it is still being investigated in some forms in different cancers and with a variety of antigens as well. One of the subsets of cancer immunotherapy is its treatment using parasitic antigens. The present study evaluated the effect of using somatic antigens of protoscoleces of Echinococcus granulosus on K562 cancer cells. METHODS In this study, hydatid cysts' protoscolex antigens were extracted, purified, and added to K562 cancer cells at three concentrations (0.1, 1, and 2 mg/ml) and on three times (24, 48, and 72 h). The number of apoptotic cells was compared to the control flask. The antigen concentration of 2 mg/ml was used as a control sample to investigate its cytotoxic effect on the growth of healthy HFF3 cells. Annexin V and PI tests were also performed to differentiate apoptosis from necrosis. RESULTS In flasks treated with hydatid cyst protoscolex antigen, all three concentrations significantly reduced the growth of cancer cells compared with the control flask, and concentration 2 of crude antigen significantly caused the death of cancer cells. Furthermore, more cancer cells underwent apoptosis by increasing the time of exposure to the antigen. On the other hand, flow cytometry results also showed that the amount of apoptosis has increased compared to the control group. In fact, Protoscolex hydatid cyst somatic antigens induce programmed cell death in K562 cancer cells while not having a cytotoxic effect on normal cells. CONCLUSION Therefore, it is suggested to do more research on the anti-cancer and therapeutic properties of the antigens of this parasite.
Collapse
Affiliation(s)
- Atefe Asouli
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Soheil Sadr
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hadi Mohebalian
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran.
| |
Collapse
|
45
|
Zinc Finger Protein 90 Knockdown Promotes Cisplatin Sensitivity via Nrf2/HO-1 Pathway in Ovarian Cancer Cell. Cancers (Basel) 2023; 15:cancers15051586. [PMID: 36900383 PMCID: PMC10000492 DOI: 10.3390/cancers15051586] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Our study discussed the role of Zfp90 in ovarian cancer (OC) cell lines' sensitivity to cisplatin. We used two OC cell lines, SK-OV-3 and ES-2, to evaluate their role in cisplatin sensitization. The protein levels of p-Akt, ERK, caspase 3, Bcl-2, Bax, E-cadherin, MMP-2, MMP-9 and other drug resistance-related molecules, including Nrf2/HO-1, were discovered in the SK-OV-3 and ES-2 cells. We also used a human ovarian surface epithelial cell to compare the effect of Zfp90. Our outcomes indicated that cisplatin treatment generates reactive oxygen species (ROS) that modulate apoptotic protein expression. The anti-oxidative signal was also stimulated, which could hinder cell migration. The intervention of Zfp90 could greatly improve the apoptosis pathway and block the migrative pathway to regulate the cisplatin sensitivity in the OC cells. This study implies that the loss of function of Zfp90 might promote cisplatin sensitization in OC cells via regulating the Nrf2/HO-1 pathway to enhance cell apoptosis and inhibit the migrative effect in both SK-OV-3 and ES-2 cells.
Collapse
|
46
|
Short peptide domains of the Wnt inhibitor sFRP4 target ovarian cancer stem cells by neutralizing the Wnt β-catenin pathway, disrupting the interaction between β-catenin and CD24 and suppressing autophagy. Life Sci 2023; 316:121384. [PMID: 36646377 DOI: 10.1016/j.lfs.2023.121384] [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: 09/29/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
AIMS One of the hallmarks of cancer stem cells (CSC) is hyperactive Wnt β-catenin signaling due to the decreased presence of Wnt antagonists such as secreted frizzled related protein 4 (SFRP4). Cysteine-rich domain (CRD) and netrin-like domain (NLD) are the two functional domains of SFRP4 having anti-tumor properties. In this study, we have explored the effectiveness of short micropeptides SC-301 (from CRD) and SC-401 (from NLD) on CSC properties, EMT, apoptosis and autophagy in ovarian CSCs enriched from PA-1 and SKOV-3 cell lines. MAIN METHODS Gene expression analysis, Western blot and immunocytochemistry were performed on ovarian CSCs to evaluate the inhibitory potential of micropeptides to various CSC associated oncogenic properties. Co-immunoprecipitation was performed to detect the binding of CD24 to β-catenin protein complex. CYTO-ID Autophagy Detection Kit 2.0 was used to monitor autophagic flux in peptide treated CSCs. KEY FINDINGS It is clearly seen that the micropeptides derived from both the domains inhibit Wnt pathway, initiate apoptosis, inhibit migration and chemosensitize CSCs. Specifically, CD24, a defining marker of ovarian CSC was suppressed by peptide treatment. Notably, interaction between CD24 and β-catenin was disrupted upon peptide treatment. SFRP4 peptide treatment also suppressed the autophagic process which is crucial for CSC survival. SIGNIFICANCE The study demonstrated that although both peptides have inhibitory effects, SC-401 was emphatically more effective in targeting CSC properties and down regulating the Wnt β-catenin machinery.
Collapse
|
47
|
Győrffy B. Discovery and ranking of the most robust prognostic biomarkers in serous ovarian cancer. GeroScience 2023:10.1007/s11357-023-00742-4. [PMID: 36856946 PMCID: PMC10400493 DOI: 10.1007/s11357-023-00742-4] [Citation(s) in RCA: 158] [Impact Index Per Article: 158.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/25/2023] [Indexed: 03/02/2023] Open
Abstract
Progress in ovarian cancer treatment lags behind other tumor types. With diagnosis usually at an advanced stage, there is a high demand for reliable prognostic biomarkers capable of the selection of effective chemo- and targeted therapies. Our goal was to establish a large-scale transcriptomic database and use it to uncover and rank survival-associated genes. Ovarian cancer cohorts with transcriptome-level gene expression data and clinical follow-up were identified from public repositories. All samples were normalized and entered into an integrated database. Cox univariate survival analysis was performed for all genes and was followed by multivariate analysis for selected genes involving clinical and pathological variables. False discovery rate was computed for multiple hypothesis testing and a 1% cutoff was used to determine statistical significance. The complete integrated database comprises 1816 samples from 17 datasets. Altogether, 2468 genes were correlated to progression-free survival (PFS), and 704 genes were correlated with overall survival (OS). The most significant genes were WBP1L, ASAP3, CNNM2, and NCAPH2 for progression-free survival and CSE1L, NUAK1, ALPK2, and SHKBP1 for overall survival. Genes significant for PFS were also preferentially significant for predicting OS as well. All data including HR and p values as well as the used cutoff values for all genes for both PFS and OS are provided to enable the ranking of future biomarker candidates across all genes. Our results help to prioritize genes and to neglect those which are most likely to fail in studies aiming to establish new clinically useful biomarkers and therapeutic targets in serous ovarian cancer.
Collapse
Affiliation(s)
- Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, Tuzolto U. 7-9, 1094, Budapest, Hungary.
| |
Collapse
|
48
|
Li Y, Huang X, Tang J. Inhibiting the growth of ovarian cancer cells in vitro and in vivo by a small molecular inhibitor targeting La-RNA interactions. Eur J Pharmacol 2023; 940:175471. [PMID: 36549502 DOI: 10.1016/j.ejphar.2022.175471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To identify small molecules blocking La-RNA interactions by using structural dynamics, molecular biology, and in vivo efficacy experiments. METHODS A docking virtual assay on the Chemdiv database was used to screen La binders, and their affinity were measured by surface plasmon resonance (SPR). A novel fluorescence polarization (FP) assay referring to the binding of La protein and 3'UUUOH was established to identify the inhibitors. Their activity on ovarian cancer cell proliferation, apoptosis and cell cycle were evaluated using Cell Counting Kit 8 (CCK8) and flow cytometry assay, respectively. Their in vivo efficacy against ovarian cancer growth were evaluated in a cell line-derived xenograft (CDX) model of A2780 cells. RESULTS From a total of 20 compounds with high potential binding activity with La protein, two small molecule compounds 4424-1120 and 8017-5932 with relatively stronger inhibition ability on La-RNA interactions were identified. These two compounds shared the same active centers with hydroxyimidazole and hydroxybenzene to interact with La protein through residues ARG57, GLN20 and GLN136. The in vitro assays showed that 4424-1120 and 8017-5932 effectively cause G0/G1 cell cycle arrest, inhibit cell proliferation, reduce cell invasion and promote apoptosis in ovarian cancer cells. In a CDX model on BALB/C Nude mice, we found that the growth rate of the tumor was inhibited by 4424-1120. CONCLUSION Our results demonstrated compound 4424-1120 shows good antitumor activity and safety in vitro and in vivo, and it provides a new idea for the discovery of antitumor lead compounds from small drug-like molecules.
Collapse
Affiliation(s)
- Yueyan Li
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| | - Xuan Huang
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| | - Jing Tang
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
49
|
Masood A, Sarfaraz R, Zaki S, Shami A, Khaliq S, Naseem N. Potential prognostic role of somatic mutations in a set of cancer susceptibility genes in ovarian carcinoma: A follow-up multicentric study from Pakistan. Cancer Biomark 2023; 36:207-219. [PMID: 36776043 DOI: 10.3233/cbm-220267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Genetic mutations, peritoneal metastasis and frequent development of chemoresistance worsen the prognosis of Ovarian carcinoma. OBJECTIVE The objective of the study is to determine mutations in cancer susceptibility genes in relation with chemotherapy response. METHODS In this follow up descriptive study, 47 consenting female patients diagnosed with surface epithelial ovarian cancer were observed for six months after completion of chemotherapy to see the treatment response. For genetic analysis, the DNA extraction was done and the genomic regions of different exons of BRCA1/2, PALB2, CHEK2, BAP1, CTNNB1, HOXB13, and PIK3CA were amplified using gene specific primers followed by Sanger Sequencing. RESULTS 86.7% of the patients were sensitive to chemotherapy whereas 13.3% showed resistance. Genetic variants of BRCA1 in 7%, BRCA2 in 4.7%, PIK3CA in 9.3%, PALB2 in 7%, CHEK2 in 2.3%, BAP1 in 2.3%, and CTNNB1 in 2.3% of the patients were found. There was also a significant association between TNM stage and the treatment response (p< 0.01). Of the patients with no mutations, 90.9% showed chemosensitivity as opposed to 70% in mutations group. CONCLUSION Our study exhibits the pivotal role of genetic analysis in predicting the treatment response and paving pathway for patient tailored targeted therapy in Pakistani population.
Collapse
Affiliation(s)
- Atika Masood
- Department of Histopathology and Morbid Anatomy, University of Health Sciences, Lahore, Pakistan
| | - Rahat Sarfaraz
- Department of Histopathology and Morbid Anatomy, University of Health Sciences, Lahore, Pakistan
| | - Saima Zaki
- Department of Obstetrics and Gynecology, Jinnah Hospital, Lahore, Pakistan
| | - Amira Shami
- Department of Oncology, INMOL Hospital, Lahore, Pakistan
| | - Saba Khaliq
- Department of Physiology and Cell Biology, University of Health Sciences, Lahore, Pakistan
| | - Nadia Naseem
- Department of Histopathology and Morbid Anatomy, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
50
|
Darroudi M, Elnaz Nazari S, Karimzadeh M, Asgharzadeh F, Khalili-Tanha N, Asghari SZ, Ranjbari S, Babaei F, Rezayi M, Khazaei M. Two-dimensional-Ti 3C 2 magnetic nanocomposite for targeted cancer chemotherapy. Front Bioeng Biotechnol 2023; 11:1097631. [PMID: 36761295 PMCID: PMC9905703 DOI: 10.3389/fbioe.2023.1097631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction: Cervical cancer is the leading cause of cancer-related death in women, so novel therapeutic approaches are needed to improve the effectiveness of current therapies or extend their activity. In recent decades, graphene analogs, such as Mxene, an emerging class of two-dimensional (2D) graphene analogs, have been drawing considerable attention based on their intrinsic physicochemical properties and performance as potential candidates for tumor therapy, particularly for therapeutic purposes. Here we explored the targeted drug delivery in cervical cancer in in vivo model. Mxene-based nanocarriers are not able to be precisely controlled in cancer treatment. Method: To solve this problem, the titanium carbide-magnetic core-shell nanocarrier (Ti3C2-Fe3O4@SiO2-FA) is also developed to provide synergetic anticancer with magnetic controlling ability along with pH-responsive drug release. A xenograft model of the cervix was used to investigate the effects of Cisplatin alone, or in combination with Ti3C2@FA and Ti3C2@ Fe3O4@SiO2-FA, on tumor growth following histological staining for evaluation of necrosis. Result and Discussion: A significant tumor-growth suppression effect is shown when the Ti3C2-Fe3O4@SiO2-FA nanocarrier is magnetically controlled Cisplatin drug release. It reveals a synergistic therapeutic efficacy used in conjunction with pharmaceuticals (p < .001). According to the in vivo study, the Ti3C2@FA@Cisplatin nanocomposite exhibits less tumor growth than the drug alone or Ti3C2@FA@Cisplatin via increasing necrosis effect (p < .001). Through this study, Mxene nanosheets are expanded for biomedical applications, not only through the fabrication of biocompatible magnetic Mxene nanocomposite but also through the development of functionalization strategies that enable the magnetic Ti3C2 nanocomposite to load high levels of Cisplatin for cervical cancer treatment (242.5%). Hence, Ti3C2-Fe3O4@SiO2-FA nanocarriers would be promising candidates to improve cancer treatment efficiency.
Collapse
Affiliation(s)
- Mahdieh Darroudi
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran,Department of Medical Biotechnology and Nanotechnology, School of Science, Mashhad University of Medical Science, Mashhad, Iran,Department of Electrical and Computer Engineering, University of Central Florida, Orlando, FL, United states
| | - Seyedeh Elnaz Nazari
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Maryam Karimzadeh
- Department of Medical Biotechnology and Nanotechnology, School of Science, Mashhad University of Medical Science, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Nima Khalili-Tanha
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Seyyedeh Zahra Asghari
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Sara Ranjbari
- Chemical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh Babaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Majid Rezayi
- Department of Medical Biotechnology and Nanotechnology, School of Science, Mashhad University of Medical Science, Mashhad, Iran,Metabolic Syndrome Research Centre, Mashhad University of Medical Science, Mashhad, Iran,Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,*Correspondence: Majid Rezayi, ; Majid Khazaei,
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran,Metabolic Syndrome Research Centre, Mashhad University of Medical Science, Mashhad, Iran,*Correspondence: Majid Rezayi, ; Majid Khazaei,
| |
Collapse
|