1
|
Li X, Wang Y, Geng X, Sun J, Liu Y, Dong A, Zhang R. Melanin-intercalated layered double hydroxide LDH/MNP as a stable photothermal agent. BMC Chem 2024; 18:198. [PMID: 39396055 PMCID: PMC11471033 DOI: 10.1186/s13065-024-01312-1] [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: 02/27/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024] Open
Abstract
Melanin nanoparticles (MNPs) are a type of electronegative compound that can be used as photothermal agent for cancer treatment. Nevertheless, the agglomeration of MNP, which is one of the limitations in practice, contributes to the instability of MNP. Pristine layered double hydroxide (LDH), as a kind of positive inorganic material when there exist no other cargo between its layers, can accommodate electronegative molecules between its layers to endow them with stable properties. Hence, in this study, electronegative MNP was intercalated into LDH lamellas via ion-exchange method to obtain the stable original photothermal agent LDH/MNP, solving the tough problem of MNP's agglomeration. The surface morphology, X-ray diffraction and fourier transform infrared spectra affirmed the successful intercalation of MNP between LDH lamellas. The Z-average particle sizes of LDH/MNP on day 0, 7 and 14 were measured as 221.8 nm, 227.6 nm and 230.5 nm without obvious fluctuation, while the particle sizes of MNP went through dramatic enlargement from 105.8 nm (day 0) to 856.1 nm (day 7), indicating the better stability of LDH/MNP than MNP. The typical polymer dispersity index (PDI) values on day 0, 7 and 14 verified the better stability of LDH/MNP, too. Photothermal properties of LDH/MNP were assessed and the results ensured the representative photothermal properties of LDH/MNP. The fine cytocompatibility of LDH/MNP was verified via cytotoxicity test. Results confirmed that the agglomeration of MNP disappeared after its intercalation into LDH and LDH/MNP possessed fine stability as well as typical photothermal property. The intercalation of MNP into LDH gave the photothermal agent MNP a promising way for its better stability and long-term availability in photothermal treatment.
Collapse
Affiliation(s)
- Xue Li
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
- Department of Chemistry, School of Basic Medicine, Shanxi Medical University, Shanxi, 030001, China
| | - Yixuan Wang
- The First Clinical Medical College of Shanxi Medical University, Shanxi, 030001, China
| | - Xinkai Geng
- The First Clinical Medical College of Shanxi Medical University, Shanxi, 030001, China
| | - Jinghua Sun
- The First Clinical Medical College of Shanxi Medical University, Shanxi, 030001, China
| | - Yulong Liu
- Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Anjie Dong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, Fifth Hospital of Shanxi Medical University, Shanxi, 030001, China.
| |
Collapse
|
2
|
De Landtsheer S, Badkas A, Kulms D, Sauter T. Model ensembling as a tool to form interpretable multi-omic predictors of cancer pharmacosensitivity. Brief Bioinform 2024; 25:bbae567. [PMID: 39494610 PMCID: PMC11532660 DOI: 10.1093/bib/bbae567] [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: 07/10/2024] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Stratification of patients diagnosed with cancer has become a major goal in personalized oncology. One important aspect is the accurate prediction of the response to various drugs. It is expected that the molecular characteristics of the cancer cells contain enough information to retrieve specific signatures, allowing for accurate predictions based solely on these multi-omic data. Ideally, these predictions should be explainable to clinicians, in order to be integrated in the patients care. We propose a machine-learning framework based on ensemble learning to integrate multi-omic data and predict sensitivity to an array of commonly used and experimental compounds, including chemotoxic compounds and targeted kinase inhibitors. We trained a set of classifiers on the different parts of our dataset to produce omic-specific signatures, then trained a random forest classifier on these signatures to predict drug responsiveness. We used the Cancer Cell Line Encyclopedia dataset, comprising multi-omic and drug sensitivity measurements for hundreds of cell lines, to build the predictive models, and validated the results using nested cross-validation. Our results show good performance for several compounds (Area under the Receiver-Operating Curve >79%) across the most frequent cancer types. Furthermore, the simplicity of our approach allows to examine which omic layers have a greater importance in the models and identify new putative markers of drug responsiveness. We propose several models based on small subsets of transcriptional markers with the potential to become useful tools in personalized oncology, paving the way for clinicians to use the molecular characteristics of the tumors to predict sensitivity to therapeutic compounds.
Collapse
Affiliation(s)
- Sébastien De Landtsheer
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Apurva Badkas
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technische Universität-Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases, Technische Universität-Dresden, 01307 Dresden, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
3
|
Adusumilli P, Ravikumar N, Hall G, Swift S, Orsi N, Scarsbrook A. Radiomics in the evaluation of ovarian masses - a systematic review. Insights Imaging 2023; 14:165. [PMID: 37782375 PMCID: PMC10545652 DOI: 10.1186/s13244-023-01500-y] [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: 05/07/2023] [Accepted: 08/12/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVES The study aim was to conduct a systematic review of the literature reporting the application of radiomics to imaging techniques in patients with ovarian lesions. METHODS MEDLINE/PubMed, Web of Science, Scopus, EMBASE, Ovid and ClinicalTrials.gov were searched for relevant articles. Using PRISMA criteria, data were extracted from short-listed studies. Validity and bias were assessed independently by 2 researchers in consensus using the Quality in Prognosis Studies (QUIPS) tool. Radiomic Quality Score (RQS) was utilised to assess radiomic methodology. RESULTS After duplicate removal, 63 articles were identified, of which 33 were eligible. Fifteen assessed lesion classifications, 10 treatment outcomes, 5 outcome predictions, 2 metastatic disease predictions and 1 classification/outcome prediction. The sample size ranged from 28 to 501 patients. Twelve studies investigated CT, 11 MRI, 4 ultrasound and 1 FDG PET-CT. Twenty-three studies (70%) incorporated 3D segmentation. Various modelling methods were used, most commonly LASSO (least absolute shrinkage and selection operator) (10/33). Five studies (15%) compared radiomic models to radiologist interpretation, all demonstrating superior performance. Only 6 studies (18%) included external validation. Five studies (15%) had a low overall risk of bias, 9 (27%) moderate, and 19 (58%) high risk of bias. The highest RQS achieved was 61.1%, and the lowest was - 16.7%. CONCLUSION Radiomics has the potential as a clinical diagnostic tool in patients with ovarian masses and may allow better lesion stratification, guiding more personalised patient care in the future. Standardisation of the feature extraction methodology, larger and more diverse patient cohorts and real-world evaluation is required before clinical translation. CLINICAL RELEVANCE STATEMENT Radiomics shows promising results in improving lesion stratification, treatment selection and outcome prediction. Modelling with larger cohorts and real-world evaluation is required before clinical translation. KEY POINTS • Radiomics is emerging as a tool for enhancing clinical decisions in patients with ovarian masses. • Radiomics shows promising results in improving lesion stratification, treatment selection and outcome prediction. • Modelling with larger cohorts and real-world evaluation is required before clinical translation.
Collapse
Affiliation(s)
- Pratik Adusumilli
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
- West Yorkshire Radiology Academy, Level B Clarendon Wing, Leeds General Infirmary, Great George Street, Leeds, LS1 3EX, UK.
| | - Nishant Ravikumar
- Centre for Computational Imaging and Simulation Technologies in Biomedicine, University of Leeds, Leeds, UK
| | - Geoff Hall
- Department of Medical Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Sarah Swift
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Nicolas Orsi
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Andrew Scarsbrook
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| |
Collapse
|
4
|
Huang ML, Ren J, Jin ZY, Liu XY, He YL, Li Y, Xue HD. A systematic review and meta-analysis of CT and MRI radiomics in ovarian cancer: methodological issues and clinical utility. Insights Imaging 2023; 14:117. [PMID: 37395888 DOI: 10.1186/s13244-023-01464-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/11/2023] [Indexed: 07/04/2023] Open
Abstract
OBJECTIVES We aimed to present the state of the art of CT- and MRI-based radiomics in the context of ovarian cancer (OC), with a focus on the methodological quality of these studies and the clinical utility of these proposed radiomics models. METHODS Original articles investigating radiomics in OC published in PubMed, Embase, Web of Science, and the Cochrane Library between January 1, 2002, and January 6, 2023, were extracted. The methodological quality was evaluated using the radiomics quality score (RQS) and Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). Pairwise correlation analyses were performed to compare the methodological quality, baseline information, and performance metrics. Additional meta-analyses of studies exploring differential diagnoses and prognostic prediction in patients with OC were performed separately. RESULTS Fifty-seven studies encompassing 11,693 patients were included. The mean RQS was 30.7% (range - 4 to 22); less than 25% of studies had a high risk of bias and applicability concerns in each domain of QUADAS-2. A high RQS was significantly associated with a low QUADAS-2 risk and recent publication year. Significantly higher performance metrics were observed in studies examining differential diagnosis; 16 such studies as well as 13 exploring prognostic prediction were included in a separate meta-analysis, which revealed diagnostic odds ratios of 25.76 (95% confidence interval (CI) 13.50-49.13) and 12.55 (95% CI 8.38-18.77), respectively. CONCLUSION Current evidence suggests that the methodological quality of OC-related radiomics studies is unsatisfactory. Radiomics analysis based on CT and MRI showed promising results in terms of differential diagnosis and prognostic prediction. CRITICAL RELEVANCE STATEMENT Radiomics analysis has potential clinical utility; however, shortcomings persist in existing studies in terms of reproducibility. We suggest that future radiomics studies should be more standardized to better bridge the gap between concepts and clinical applications.
Collapse
Affiliation(s)
- Meng-Lin Huang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Ren
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xin-Yu Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yong-Lan He
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China.
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, People's Republic of China.
| | - Hua-Dan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
5
|
Yang B, Yin S, Zhou Z, Huang L, Xi M. Inflammation Control and Tumor Growth Inhibition of Ovarian Cancer by Targeting Adhesion Molecules of E-Selectin. Cancers (Basel) 2023; 15:cancers15072136. [PMID: 37046797 PMCID: PMC10093113 DOI: 10.3390/cancers15072136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Objective: The aim is to use E-selectin-binding peptide (ESBP) to actively recognize E-selectin, so allowing a drug delivery system to actively recognize the cells and inhibit the tumor growth of ovarian cancer by targeting adhesion molecules of E-selectin. An ovarian-cancer-directed drug delivery system was designed based on the high affinity of E-selectin-binding peptide (ESBP) to E-selectin. The effects and mechanisms of ESBP-bovine serum albumin (BSA) polymerized nanoparticles were investigated. Methods: BSA polymerized nanoparticles (BSANPs) and ESBP-BSANPs-paclitaxel (PTX) were prepared and their characteristics were measured. The in vitro targetability and cytotoxicity of ESBP-BSANPs-PTX were evaluated through in vitro drug uptake and MTT experiments. The mechanisms of ESBP-BSANPs-PTX were investigated via apoptosis, wound healing and immunohistochemistry assays. The in vivo targeting properties and drug effects were observed in a mouse tumor-bearing model. Results: In vitro experiments revealed an increase in the uptake of ESBP-BSANPs-FITC. The cytotoxicity of ESBP-BSANPs-PTX in A2780/CP70, HUVEC, RAW264.7 and ID8 cells was higher than that of PTX alone. ESBP-BSANPs-PTX increased cell apoptosis in a dose-dependent manner and exhibited a greater ability to inhibit cell migration than BSANPs-PTX. In vivo experiments demonstrated the targetability and good effects of ESBP-BSANPs. Conclusions: ESBP-BSANPs-PTX improve PTX targetability, provide tumor-specific and potent therapeutic activities, and show promise for the development of agents in preclinical epithelial ovarian cancer.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Shanmei Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zishuo Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Luyao Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Mingrong Xi
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
6
|
Kotaniemi‐Talonen L, Pukkala E, Aittomäki K, Auranen A. Long-term risk of cancer among the first-degree relatives of epithelial ovarian cancer patients: A cohort study with 48 years of follow up. Acta Obstet Gynecol Scand 2023; 102:240-245. [PMID: 36645194 PMCID: PMC9951276 DOI: 10.1111/aogs.14504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The long-term risk of cancer among first-degree relatives of ovarian cancer patients, especially their offspring, is of apparent clinical importance. Risks caused by known inherited factors such as BRCA1 or BRCA2 pathogenic variants are well established, but these account for only about 15% of ovarian cancer cases. Less is known about the possible familial risks of sporadic ovarian cancers. MATERIAL AND METHODS Using registry data, we conducted a retrospective cohort study with a total of 6501 first-degree relatives of 559 epithelial ovarian cancer patients. We studied the occurrence of overall cancer and cancer in specific sites known or suspected to be associated with ovarian cancer (breast, cervix, colon, endometrium, lung and trachea, skin melanoma, ovary, pancreas, prostate, rectum, and stomach). RESULTS The overall number of cancers was not increased among the first-degree relatives of epithelial ovarian cancer patients during the up to 48 years of follow up. Among female relatives, the standardized incidence ratio for ovarian cancer was 1.92 (95% CI 1.27-2.79), mostly explained by a 2.30-fold (95% CI 1.46-3.45) risk among the patients' sisters. There was a decreasing trend in the standardized incidence ratio for ovarian cancer among patients' sisters by increasing age of the index patient. CONCLUSIONS In our study cohort, we did not observe an increase in the overall cancer risk among the first-degree relatives of epithelial ovarian cancer patients in comparison with the general population. The risk for ovarian cancer, however, was increased. Current recommendations suggest prophylactic removal of the fallopian tubes and ovaries only with identified inherited risk factors. Our results emphasize the role of genetic counseling and testing, particularly in young ovarian cancer patients and their close female relatives.
Collapse
Affiliation(s)
- Laura Kotaniemi‐Talonen
- Department of Obstetrics and GynecologyTampere University HospitalTampereFinland,Tays Cancer CentreTampere University HospitalTampereFinland,Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Eero Pukkala
- Finnish Cancer Registry—Institute for Statistical and Epidemiological Cancer ResearchHelsinkiFinland,Health Sciences Unit, Faculty of Social SciencesTampere UniversityTampereFinland
| | - Kristiina Aittomäki
- Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Annika Auranen
- Department of Obstetrics and GynecologyTampere University HospitalTampereFinland,Tays Cancer CentreTampere University HospitalTampereFinland,Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| |
Collapse
|
7
|
Zhou Z, Jin L, Shen J, Shi W, Xu Y, Ye L, Liu J, Pan J. COM33 suppresses carboplatin-induced epithelial-mesenchymal transition via inhibition of Twist1 in ovarian cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 55:34-42. [PMID: 36647720 PMCID: PMC10157527 DOI: 10.3724/abbs.2022195] [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: 10/28/2021] [Accepted: 05/08/2022] [Indexed: 12/23/2022] Open
Abstract
Despite favorable responses to platinum-based chemotherapy in ovarian cancer (OC), chemoresistance is still a major cause of treatment failure. Hence, we develop a novel synthetic agent, COM33, to relieve the chemoresistance caused by carboplatin. The anti-cancerous effects of the combination of COM33 and carboplatin on OC are evaluated by cell viability, wound healing, and transwell invasion assays. A mechanistic investigation is carried out by using RNA-Seq analysis and then verified by western blot analysis and immunofluorescence microscopy. The safety and efficacy in vivo are evaluated using SKOV3 tumor-bearing nude mice. Results show that the co-administration of COM33 enhances the inhibitory effects of carboplatin on cancer cell viability, migration, and invasion in vitro and tumor growth in vivo. Furthermore, COM33 suppresses the carboplatin-induced epithelial-mesenchymal transition (EMT) by inhibiting the ERK signaling pathway. Additionally, we show that Twist1, the effector of the ERK signaling pathway, participates in carboplatin-induced EMT and is also inhibited by COM33. Our data show that the combination of carboplatin with COM33 is beneficial for chemotherapy against OC, which may be a potential novel anti-tumor strategy.
Collapse
Affiliation(s)
- Zhiyang Zhou
- Obstetrics & Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200011China
| | - Li Jin
- Obstetrics & Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200011China
| | - Jian Shen
- College of Life ScienceZhejiang Chinese Medical UniversityHangzhou310053China
| | - Weihui Shi
- Obstetrics & Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200011China
| | - Yue Xu
- Obstetrics & Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200011China
| | - Longyun Ye
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Junxi Liu
- Chinese Academy of Science Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu ProvinceLanzhou Institute of Chemical PhysicsChinese Academy of SciencesLanzhou730000China
| | - Jiexue Pan
- Obstetrics & Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200011China
| |
Collapse
|
8
|
DNA methylation of the immediate upstream region of BRCA1 major transcription start sites is an independent favorable prognostic factor in patients with high-grade serous ovarian cancer. Gynecol Oncol 2022; 167:513-518. [PMID: 36253303 DOI: 10.1016/j.ygyno.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To establish a quantitative method to evaluate the DNA methylation level of an immediate upstream region of major BRCA1 transcriptional start sites (TSSs), and to investigate whether methylation of the region is a prognostic factor in high-grade serous ovarian cancer patients after neoadjuvant chemotherapy. METHODS Ninety-two FFPE samples of advanced high-grade serous ovarian cancers after neoadjuvant chemotherapy between 2011 and 2018 were used for mutation and methylation analysis. DNA methylation levels were assessed by pyrosequencing and DNA methylation microarray. An association between methylation level (or a mutation) and progression-free survival was assessed by Kaplan-Meier analysis. RESULT Major BRCA1 transcripts and CpG sites immediately upstream of their TSSs were identified, and a pyrosequencing method was developed. BRCA1 methylation, BRCA1/2 mutations, and a RAD51C mutation were detected in 17/79 (21.5%), 17/92 (18.5%), and 1/92 (1.1%) high-grade serious ovarian cancer samples. In univariate analysis, BRCA1 methylation and no residual tumor were associated with progression-free survival (BRCA1 methylation: P = 0.025, no residual tumor: P = 0.0026). Multivariate analysis showed that both BRCA1 methylation (P = 0.038, HR = 0.47, 95% CI: 0.21-0.96) and no residual tumor (P = 0.012, HR = 0.49, 95% CI: 0.28-0.85) were significant favorable prognostic factors. CONCLUSION A quantitative method to estimate the methylation level of the immediate upstream region of major BRCA1 TSSs was established. Methylation of the region of was an independent favorable prognostic factor in high-grade serous ovarian cancer patients.
Collapse
|
9
|
Xing Y, Liang X, Lv X, Cheng Y, Du J, Liu C, Yang Y. New insights into the role of circular RNAs in ovarian cancer. Pathol Res Pract 2022; 238:154073. [PMID: 36007396 DOI: 10.1016/j.prp.2022.154073] [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: 04/12/2022] [Revised: 07/31/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Ovarian cancer (OC) is one of the most aggressive tumors in women and has a poor prognosis and the highest mortality rate. Circular RNAs (circRNAs) are a type of endogenous non-coding RNAs that have recently attracted interest in cancer research. Increasing evidence has demonstrated that circRNAs play an oncogenic or suppressive role in tumorigenesis and progression, and show tissue- or developmental-stage-specific expression. Due to high stability, conservation, abundance, and specificity, circRNAs are considered promising biomarkers for the diagnosis and prognosis of cancer. Herein, we have summarized the expression profiles of circRNAs in OC tissues, serums, and cell lines. Moreover, we discuss how circRNAs participate in the regulation of multiple biological processes in OC, including cell proliferation, apoptosis, migration, invasion, autophagy, epithelial-to-mesenchymal transition, glucose metabolism, angiogenesis, immune response, and chemotherapy resistance, by sponging microRNAs and interacting with proteins.
Collapse
Affiliation(s)
- Yijuan Xing
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou 730000 Gansu, China
| | - Xiao Lv
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou 730000 Gansu, China
| | - Yuemei Cheng
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000 Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou 730000 Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou 730000, Gansu, China; Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou 730000 Gansu, China.
| |
Collapse
|
10
|
Li R, Li H, Lan J, Yang D, Lin X, Xu H, Han B, Yang M, Su B, Liu F, Jiang W. Damnacanthal isolated from morinda species inhibited ovarian cancer cell proliferation and migration through activating autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154084. [PMID: 35421676 DOI: 10.1016/j.phymed.2022.154084] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/28/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Ovarian cancer is a very common gynecological malignant tumor. Natural products are important sources of chemotherapy drugs for ovarian cancer. Damnacanthal is an anthraquinone derivative with anti-cancer pharmacological properties. OBJECTIVE This study aimed to investigate the mechanisms underlying damnacanthal's effects against ovarian cancer. METHODS In vitro experiments, CCK8, colony formation and flow cytometry assays were used to evaluate the anti-ovarian cancer effect of damnacanthal on SKVO3 and A2780 cells. The wound healing tests and the transwell invasion assays were used to detect the migration and infiltration of ovarian cancer cells. Western Blot assays and immunofluorescence staining were used to measure autophagy levels. In vivo experiments, the anti-ovarian cancer effect of damnacanthal was further evaluated in a xenograft nude mouse model of SKVO3 cells. RESULTS Damnacanthal induced significant cell death and apoptosis, as well as significant inhibition in migration and invasion, in SKVO3 and A2780 cells, Furthermore, damnacanthal induced cell cycle arrest by increasing the protein levels of p27Kip1 and decreasing cyclin D1 levels. In addition, damnacanthal induced a significant accumulation of autophagosomes, accompanied with an increase in LC3II protein levels, and a decrease in p62 protein levels. 3-methyladenine, an autophagy formation inhibitor, significantly mitigated the damnacanthal-induced apoptosis and migration hindrance, as well as the decline in cell viability. Furthermore, the inactivation of ERK and its downstream effector mTOR signaling pathways, rather than Akt or P38 pathway, were involved in damnacanthal's activation in autophagy. In addition, TBHQ, an ERK activator, significantly inhibited damnacanthal-boosted LC3 II levels and autophagosome accumulation, and reversed damnacanthal-induced cell death, apoptosis, cell cycle arrest and migration hindrance. Finally, the anti-ovarian cancer effect of damnacanthal was confirmed in the orthotopic xenograft model of SKVO3 cells in nude mice, with tumor growth being significantly inhibited comparably to the efficacy of cisplatin. Damnacanthal was also synergistic with cisplatin and showed inhibition in cisplatin-resistant ovarian cancer cells. CONCLUSION Damnacanthal inhibited the growth of ovarian cancer via the ERK/mTOR/autophagy signaling cascade, indicating that it may be a potential anti-ovarian cancer drug candidate.
Collapse
Affiliation(s)
- Ruli Li
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - He Li
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jie Lan
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dongmei Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Xinjing Lin
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Hongling Xu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Bin Han
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Bo Su
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Fu Liu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Wei Jiang
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
11
|
Mistarz A, Graczyk M, Winkler M, Singh PK, Cortes E, Miliotto A, Liu S, Long M, Yan L, Stablewski A, O'Loughlin K, Minderman H, Odunsi K, Rokita H, McGray AJR, Zsiros E, Kozbor D. Induction of cell death in ovarian cancer cells by doxorubicin and oncolytic vaccinia virus is associated with CREB3L1 activation. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:38-50. [PMID: 34632049 PMCID: PMC8479291 DOI: 10.1016/j.omto.2021.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/24/2021] [Indexed: 12/12/2022]
Abstract
We have demonstrated that oncolytic vaccinia virus synergizes with doxorubicin (DOX) in inducing immunogenic cell death in platinum-resistant ovarian cancer cells and increases survival in syngeneic and xenograft tumor models. However, the mechanisms underlying the virus- and doxorubicin-mediated cancer cell death remain unknown. In this study, we investigated the effect of the oncolytic virus and doxorubicin used alone or in combination on activation of the cytoplasmic transcription factor CREB3L1 (cyclic AMP [cAMP] response element-binding protein 3-like 1) in ovarian cancer cell lines and clinical specimens. We demonstrated that doxorubicin-mediated cell death in ovarian cancer cell lines was associated with nuclear translocation of CREB3L1 and that the effect was augmented by infection with oncolytic vaccinia virus or treatment with recombinant interferon (IFN)-β used as a viral surrogate. This combination treatment was also effective in mediating nuclear translocation of CREB3L1 in cancer cells isolated from ovarian tumor biopsies at different stages of disease progression. The measurement of CREB3L1 expression in clinical specimens of ovarian cancer revealed lack of correlation with the stage of disease progression, suggesting that understanding the mechanisms of nuclear accumulation of CREB3L1 after doxorubicin treatment alone or in combination with oncolytic virotherapy may lead to the development of more effective treatment strategies against ovarian cancer.
Collapse
Affiliation(s)
- Anna Mistarz
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Matthew Graczyk
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Marta Winkler
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Prashant K Singh
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eduardo Cortes
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Anthony Miliotto
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aimee Stablewski
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kieran O'Loughlin
- Department of Flow and Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hans Minderman
- Department of Flow and Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hanna Rokita
- Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
| | - A J Robert McGray
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Danuta Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
12
|
Li G, Gong J, Cao S, Wu Z, Cheng D, Zhu J, Huang X, Tang J, Yuan Y, Cai W, Zhang H. The Non-Coding RNAs Inducing Drug Resistance in Ovarian Cancer: A New Perspective for Understanding Drug Resistance. Front Oncol 2021; 11:742149. [PMID: 34660304 PMCID: PMC8514763 DOI: 10.3389/fonc.2021.742149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
Ovarian cancer, a common malignant tumor, is one of the primary causes of cancer-related deaths in women. Systemic chemotherapy with platinum-based compounds or taxanes is the first-line treatment for ovarian cancer. However, resistance to these chemotherapeutic drugs worsens the prognosis. The underlying mechanism of chemotherapeutic resistance in ovarian cancer remains unclear. Non-coding RNAs, including long non-coding RNAs, microRNAs, and circular RNAs, have been implicated in the development of drug resistance. Abnormally expressed non-coding RNAs can promote ovarian cancer resistance by inducing apoptosis inhibition, protective autophagy, abnormal tumor cell proliferation, epithelial-mesenchymal transition, abnormal glycolysis, drug efflux, and cancer cell stemness. This review summarizes the role of non-coding RNAs in the development of chemotherapeutic resistance in ovarian cancer, including their mechanisms, targets, and potential signaling pathways. This will facilitate the development of novel chemotherapeutic agents that can target these non-coding RNAs and improve ovarian cancer treatment.
Collapse
Affiliation(s)
- Gaofeng Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jun Gong
- Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Shulong Cao
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Zhaoyang Wu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Dong Cheng
- Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Zhu
- Hubei Enshi College, Enshi, China
| | - Xuqun Huang
- Department of Thoracic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Jingyi Tang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yuning Yuan
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Wenqi Cai
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
13
|
Huang B, Wei M, Hong L. Long noncoding RNA HULC contributes to paclitaxel resistance in ovarian cancer via miR-137/ITGB8 axis. Open Life Sci 2021; 16:667-681. [PMID: 34250246 PMCID: PMC8253452 DOI: 10.1515/biol-2021-0058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 04/01/2021] [Accepted: 04/22/2021] [Indexed: 01/05/2023] Open
Abstract
Long noncoding RNA (lncRNA) highly upregulated in liver cancer (HULC) has been reported to be implicated in chemoresistance. However, the potential mechanism of HULC in paclitaxel (PTX)-resistant ovarian cancer (OC) remains undefined. The expression of RNAs and proteins was measured by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and Western blot assay. The PTX resistance and apoptotic rate were assessed via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry, respectively. Furthermore, the interaction between miR-137 and HULC or integrin beta-8 (ITGB8) was predicted by miRcode and starBase v2.0 and then verified by dual luciferase reporter and RNA pull-down assays. In addition, the xenograft mice model was established to explore the effects of HULC in vivo. HULC was significantly upregulated and miR-137 was downregulated in PTX-resistant OC tissues and cells. Also, the HULC depletion suppressed tumor growth and PTX resistance in PTX-treated mice. miR-137 was verified as a target of HULC and directly targeted ITGB8. And HULC knockdown downregulated ITGB8 expression by targeting miR-137. miR-137 inhibitor or ITGB8 overexpression mitigated the suppressive impacts of HULC knockdown on PTX resistance. Collectively, HULC modulated ITGB8 expression to promote PTX resistance of OC by sponging miR-137.
Collapse
Affiliation(s)
- Bo Huang
- Department of Gynaecology and Obstetrics, Hubei General Hospital, No. 99 Zhang-Zhi-Dong Street, Wuchang District, Wuhan 430000, Hubei, China
| | - Min Wei
- Department of Gynaecology and Obstetrics, Hubei General Hospital, No. 99 Zhang-Zhi-Dong Street, Wuchang District, Wuhan 430000, Hubei, China
| | - Li Hong
- Department of Gynaecology and Obstetrics, Hubei General Hospital, No. 99 Zhang-Zhi-Dong Street, Wuchang District, Wuhan 430000, Hubei, China
| |
Collapse
|
14
|
Nougaret S, McCague C, Tibermacine H, Vargas HA, Rizzo S, Sala E. Radiomics and radiogenomics in ovarian cancer: a literature review. Abdom Radiol (NY) 2021; 46:2308-2322. [PMID: 33174120 DOI: 10.1007/s00261-020-02820-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/01/2020] [Accepted: 10/10/2020] [Indexed: 01/25/2023]
Abstract
Ovarian cancer remains one of the most lethal gynecological cancers in the world despite extensive progress in the areas of chemotherapy and surgery. Many studies have postulated that this is because of the profound heterogeneity that underpins response to therapy and prognosis. Standard imaging evaluation using CT or MRI does not take into account this tumoral heterogeneity especially in advanced stages with peritoneal carcinomatosis. As such, newly emergent fields in the assessment of tumor heterogeneity have been proposed using radiomics to evaluate the whole tumor burden heterogeneity as opposed to single biopsy sampling. This review provides an overview of radiomics, radiogenomics, and proteomics and examines the use of these newly emergent fields in assessing tumor heterogeneity and its implications in ovarian cancer.
Collapse
Affiliation(s)
- S Nougaret
- IRCM, Montpellier Cancer Research Institute, INSERM, U1194, University of Montpellier, 208 Ave des Apothicaires, 34295, Montpellier, France. .,Department of Radiology, Montpellier Cancer institute, 208 Ave des Apothicaires, 34295, Montpellier, France.
| | - Cathal McCague
- Department of Radiology, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Hichem Tibermacine
- IRCM, Montpellier Cancer Research Institute, INSERM, U1194, University of Montpellier, 208 Ave des Apothicaires, 34295, Montpellier, France.,Department of Radiology, Montpellier Cancer institute, 208 Ave des Apothicaires, 34295, Montpellier, France
| | - Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Stefania Rizzo
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900, Lugano, CH, Switzerland.,Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Lugano, CH, Switzerland
| | - E Sala
- Department of Radiology, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| |
Collapse
|
15
|
Ye X, An L, Wang X, Zhang C, Huang W, Sun C, Li R, Ma H, Wang H, Gao M. ALOX5AP Predicts Poor Prognosis by Enhancing M2 Macrophages Polarization and Immunosuppression in Serous Ovarian Cancer Microenvironment. Front Oncol 2021; 11:675104. [PMID: 34094977 PMCID: PMC8172172 DOI: 10.3389/fonc.2021.675104] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Background Serous ovarian cancer (SOC) is a highly lethal gynecological malignancy with poor prognosis. Given the importance of the immune-related tumor microenvironment (TME) in ovarian cancer, investigating tumor-immune interactions and identifying novel prognostic and therapeutic targets in SOC is a promising avenue of research. ALOX5AP (Arachidonate 5-Lipoxygenase Activating Protein) is a key enzyme in converting arachidonic acid to leukotriene: a crucial immune-modulating lipid mediator. However, the role of ALOX5AP in SOC has yet to be studied. Methods ALOX5AP expression patterns across ovarian cancer and their normal tissue counterparts were cross-checked using public microarray and RNA-seq analyses and then validated in clinical samples by qRT-PCR. Kaplan-Meier survival analysis was performed in multiple independent SOC patient cohorts. Univariate and multivariate Cox regression analysis were then employed to identify clinical risk parameters associated with survival, and a genomic-clinicopathologic nomogram was built. Gene enrichment, immune infiltration, and immunosuppressor correlation analyses were then evaluated. Results ALOX5AP mRNA levels in SOC tissues were significantly upregulated compared to normal tissues. Elevated ALOX5AP was markedly associated with poor overall survival and progression-free survival in multiple SOC patient cohorts as well as with adverse clinicopathological features, including lymphatic invasion, unsatisfactory cytoreductive surgery, rapid relapse after primary treatment, and platinum non-responsiveness. A predictive nomogram, which integrated ALOX5AP expression and two independent prognosis factors (primary therapy outcome and tumor residual), was conducted to predict the 3-year and 5-year survival rate of SOC patients. Mechanistically, functional and pathway enrichment analyses revealed that ALOX5AP was primarily involved in immune response and regulation. Further exploration demonstrated that ALOX5AP was highly expressed in the immunoreactive subtype of ovarian cancer and closely related to immunocyte infiltration, especially M2 macrophage polarization. Additionally, ALOX5AP was enriched in the C4 (lymphocyte depleted) immune subtype of SOC and associated with crucial immune-repressive receptors in the tumor microenvironment at the genomic level. Conclusions ALOX5AP expression indicates a worse survival outcome and has the potential to be utilized as a prognostic predictor for SOC patients. Given the availability of well-studied ALOX5AP inhibitors, this study has immediate clinical implications for the exploitation of ALOX5AP as an immunotherapeutic target in SOC.
Collapse
Affiliation(s)
- Xiang Ye
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Limei An
- Health Management Division, Rizhao Central Hospital, Rizhao, China
| | - Xiangxiang Wang
- Department of Obstetrics and Gynecology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chenyi Zhang
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Wenqian Huang
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Chenggong Sun
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Rongrong Li
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Ma
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Hongyan Wang
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Min Gao
- Department of Obstetrics and Gynecology, Gynecology Oncology Key Laboratory, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
16
|
HDAC6 Degradation Inhibits the Growth of High-Grade Serous Ovarian Cancer Cells. Cancers (Basel) 2020; 12:cancers12123734. [PMID: 33322608 PMCID: PMC7762972 DOI: 10.3390/cancers12123734] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The objective of this study was, firstly, to investigate the relationship between Histone deacetylase 6 (HDAC6) expression and survival in patients with ovarian cancer and, secondly, to test the effects of histone deacetylase 6 (HDAC6) inhibition on ovarian cancer cells in vitro. A meta-analysis of the correlation between HDAC6 gene expression and survival was performed on 3573 ovarian tumors from 19 datasets showed that high HDAC6 gene expression was associated with a decreased risk of death. Knockdown of HDAC6 gene expression with small interfering RNA (siRNA) and protein expression with a HDAC6 targeting protein degrader decreased ovarian cell proliferation, migration, and viability. Conversely, the selective inhibition of HDAC6 catalytic activity did not produce a robust inhibition of HDAC6 protein function. In summary, we demonstrated, for the first time, that HDAC6 over-expression in ovarian cancers is a favorable prognostic marker. We provide evidence to suggest that inhibition of HDAC6 catalytic activity has limited efficacy as a monotherapy in ovarian cancers. Abstract Histone deacetylase 6 (HDAC6) is a unique histone deacetylating enzyme that resides in the cell cytoplasm and is linked to the modulation of several key cancer related responses, including cell proliferation and migration. The promising anti-cancer response of the first-generation HDAC6 catalytic inhibitors continues to be assessed in clinical trials, although its role in high grade serous ovarian cancer is unclear. This study investigated HDAC6 tumor expression by immunohistochemistry in high-grade serous ovarian cancer (HGSOC) tissue samples and a meta-analysis of HDAC6 gene expression in ovarian cancer from publicly available data. The pharmacological activity of HDAC6 inhibition was assessed in a patient-derived model of HGSOC. HDAC6 was found to be highly expressed in HGSOC tissue samples and in the patient-derived HGSOC cell lines where higher HDAC6 protein and gene expression was associated with a decreased risk of death (hazard ratio (HR) 0.38, (95% confidence interval (CI), 0.16–0.88; p = 0.02); HR = 0.88 (95% CI, 0.78–0.99; p = 0.04)). Similarly, the multivariate analysis of HDAC6 protein expression, adjusting for stage, grade, and cytoreduction/cytoreductive surgery was associated with a decreased risk of death (HR = 0.19 (95% CI, 0.06–0.55); p = 0.002). Knock-down of HDAC6 gene expression with siRNA and protein expression with a HDAC6 targeting protein degrader decreased HGSOC cell proliferation, migration, and viability. Conversely, the selective inhibition of HDAC6 with the catalytic domain inhibitor, Ricolinostat (ACY-1215), inhibited HDAC6 deacetylation of α-tubulin, resulting in a sustained accumulation of acetylated α-tubulin up to 24 h in HGSOC cells, did not produce a robust inhibition of HDAC6 protein function. Inhibition of HGSOC cell proliferation by ACY-1215 was only achieved with significantly higher and non-selective doses of ACY-1215. In summary, we demonstrated, for the first time, that HDAC6 over-expression in HGSOC and all ovarian cancers is a favorable prognostic marker. We provide evidence to suggest that inhibition of HDAC6 catalytic activity with first generation HDAC6 inhibitors has limited efficacy as a monotherapy in HGSOC.
Collapse
|
17
|
Bi YN, Guan JP, Wang L, Li P, Yang FX. Clinical significance of microRNA-125b and its contribution to ovarian carcinogenesis. Bioengineered 2020; 11:939-948. [PMID: 32842846 PMCID: PMC8291798 DOI: 10.1080/21655979.2020.1814660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The underlying mechanisms of recurrence and metastasis of epithelial ovarian cancer (EOC) are largely unknown. In the present study, we investigated the clinical significance of microRNA-125b (miR-125b) and its role in ovarian tumorigenesis and progression. Seventy patients of EOC and paired tissues were enrolled from 2015 to 2017. qRT-PCR was used to evaluate miR-125b expression in tumor tissues and EOC cell line. Gain-and-loss function of miR-125b was achieved to explore the changes in cell biological function. We found that miR-125b expression in EOC tissues, especially in the high-grade tissues (P < 0.001), was significantly lower compared to the matched adjacent noncancerous tissues and associated with pathological type, stage, and overall survival (P < 0.05). Upregulation of miR-125b promoted apoptosis and decreased cell survival rate and migration, and vice versa in vitro. Mechanistically, miR-125b negatively regulated S100A4, a metastasis-associated protein. MiR-125b overexpression significantly decreased tumor growth and inhibited lung metastasis in vivo. Our results supported that miR-125b contributes to the progression of EOC by targeting S100A4. It potentially acts as a potential biomarker and therapeutic target of EOC.
Collapse
Affiliation(s)
- Ya-Nan Bi
- Department of Operating Room, The Affiliated Hospital of Qingdao University , Qingdao, Shandong, China
| | - Jin-Ping Guan
- Department of Surgery, The Affiliated Hospital of Qingdao University , Qingdao, Shandong, China
| | - Liming Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University , Qingdao, Shandong, China
| | - Ping Li
- Department of Ultrasound, The Affiliated Hospital of Qingdao University , Huangdao, Shandong, China
| | - Feng-Xia Yang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University , Huangdao, Shandong, China
| |
Collapse
|
18
|
Mullany S, Miller DS, Robison K, Levinson K, Lee YC, Yamada SD, Walker J, Markman M, Marin A, Mast P, diZerega G. Phase II study of intraperitoneal submicron particle paclitaxel (SPP) plus IV carboplatin and paclitaxel in patients with epithelial ovarian cancersurgery. Gynecol Oncol Rep 2020; 34:100627. [PMID: 32953961 PMCID: PMC7486435 DOI: 10.1016/j.gore.2020.100627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 01/30/2023] Open
Abstract
Patients with ovarian cancer were treated with intraperitoneal submicron particle paclitaxel after debulking surgery. Following surgery, patients received IV chemotherapy without evidence of enhanced systemic toxicity. By RECIST 1.1 criteria, 66% of patients had progression free survival at 6 months and 1-year following surgery.
Submicron particles (~800 nm) of paclitaxel (SPP) contain 1–2 billion molecules of pure drug that release tumoricidal levels of paclitaxel over many weeks. This study compared two dose-levels of SPP instilled into the peritoneal cavity (IP) in 200 ml of saline post-cytoreductive surgery. Eligible patients with primary (n = 6) or recurrent (n = 4) epithelial ovarian cancer who underwent complete cytoreductive surgery were enrolled to receive a single instillation of IP SPP followed by standard IV carboplatin and paclitaxel. Endpoints were PFS and evaluation of treatment emergent adverse events. Clinical response was determined by symptoms, physical exams, CT scans, and serum CA-125 measurements. Of the 24 subjects screened, 10 were enrolled and received treatment: seven patients received 100 mg/m2 and three received 200 mg/m2. Seven subjects completed the 12-month follow-up period. Six patients were evaluable due to one subject who had unevaluable scans throughout the follow-up period and was thus excluded from PFS determination. Upon completion of planned chemotherapy post-SPP instillation, the PFS at 6 months was 66% (4/6) and at 12-months 66% (4/6) using RECIST 1.1. One subject had a complete response at the end of IV treatment but died (unrelated to study treatment) before PFS evaluation. There was one case of incision dehiscence and one case of vaginal cuff leakage after surgery. This pilot study supports further evaluation of IP SPP to treat peritoneal carcinomas.
Collapse
Affiliation(s)
- Sally Mullany
- University of Minnesota Medical Center, 2450 Riverside Ave, Minneapolis, MN 55454, USA
| | - David Scott Miller
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Katina Robison
- Women & Infants Hospital, 101 Dudley St, Providence, RI 02905, USA
| | - Kimberly Levinson
- Johns Hopkins Gynecologic Oncology at GBMC, 6569 Charles St #306, Towson, MD 21204, USA
| | - Yi-Chun Lee
- SUNY DownState Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - S Diane Yamada
- University of Chicago Medical Center, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | - Joan Walker
- University of Oklahoma, Stephenson Cancer Center, 800 NE 10th St, Oklahoma City, OK 73104, USA
| | - Maurie Markman
- Cancer Treatment Centers of America, 1331 E Wyoming Ave, Philadelphia, PA 19124, USA
| | - Alyson Marin
- US Biotest Inc., 231 Bonetti Dr # 240, San Luis Obispo, CA 93401, USA
| | - Peter Mast
- US Biotest Inc., 231 Bonetti Dr # 240, San Luis Obispo, CA 93401, USA
| | - Gere diZerega
- US Biotest Inc., 231 Bonetti Dr # 240, San Luis Obispo, CA 93401, USA.,NanOlogy, 3909 Hulen St, Fort Worth, TX 76107, USA
| |
Collapse
|
19
|
Lara OD, Bayraktar E, Amero P, Ma S, Ivan C, Hu W, Wang Y, Mangala LS, Dutta P, Bhattacharya P, Ashizawa AT, Lopez-Berestein G, Rodriguez-Aguayo C, Sood AK. Therapeutic efficacy of liposomal Grb2 antisense oligodeoxynucleotide (L-Grb2) in preclinical models of ovarian and uterine cancer. Oncotarget 2020; 11:2819-2833. [PMID: 32754300 PMCID: PMC7381098 DOI: 10.18632/oncotarget.27667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/15/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Adaptor proteins such as growth factor receptor-bound protein-2 (Grb2) play important roles in cancer cell signaling. In the present study, we examined the biological effects of liposomal antisense oligodeoxynucleotide that blocks Grb2 expression (L-Grb2) in gynecologic cancer models. MATERIALS AND METHODS Murine orthotopic models of ovarian (OVCAR5 and SKOV3ip1) and uterine (Hec1a) cancer were used to study the biological effects of L-Grb2 on tumor growth. In vitro experiments (cell viability assay, Western blot analysis, siRNA transfection, and reverse phase protein array) were carried out to elucidate the mechanisms and potential predictors of tumor response to L-Grb2. FINDINGS Treatment with L-Grb2 decreased tumor growth and metastasis in orthotopic models of ovarian cancer (OVCAR5, SKOV3ip1) by reducing angiogenesis and increasing apoptosis at a dose of 15 mg/kg with no effect on mouse body weight. Treatment with L-Grb2 and paclitaxel led to the greatest decrease in tumor weight (mean ± SEM, 0.17 g ± 0.10 g) compared with that in control mice (0.99 g ± 0.35 g). We also observed a reduction in tumor burden after treatment with L-Grb2 and the anti-VEGF antibody B-20 (86% decrease in tumor weight compared with that in controls). Ovarian cancer cells with ErbB2 amplification (OVCAR8 and SKOV3ip1) were the most sensitive to Grb2 downregulation. Reverse phase protein array analysis identified significant dysregulation of metabolites (LDHA, GAPDH, and TCA intermediates) in ovarian cancer cells after Grb2 downregulation. INTERPRETATION L-Grb2 has therapeutic efficacy in preclinical models of ovarian and uterine cancer. These findings support further clinical development of L-Grb2.
Collapse
Affiliation(s)
- Olivia D. Lara
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratip Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
|
21
|
Zhang J, Zhang Q, Zhang J, Wang Q. Expression of ACAP1 Is Associated with Tumor Immune Infiltration and Clinical Outcome of Ovarian Cancer. DNA Cell Biol 2020; 39:1545-1557. [PMID: 32456571 DOI: 10.1089/dna.2020.5596] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
ADP-ribosylation factor (Arf) GTPase-activating protein (GAP) with coiled-coil, ankyrin repeat and PH domains 1 (ACAP1) is an Arf6 GAP that regulates membrane trafficking and is critical for the migratory potential of cells. However, the roles of ACAP1 have not been fully explored and its association with clinicopathological features in ovarian cancer is still unknown. In this study, we systematically analyzed multiple databases, including TISIDB, Tumor Immune Estimation Resource (TIMER2.0), Gene Expression Omnibus (GEO), CORTECON, Kaplan-Meier Plotter and LinkedOmics platforms to reveal the clinical significance and function of ACAP1 in ovarian cancer. We found that the expression of ACAP1 was upregulated in ovarian cancer and high ACAP1 expression predicted poor prognosis. Our data demonstrated that the expression and methylation status of ACAP1 were strongly correlated with immune infiltration levels, immunomodulators, and chemokines. Gene set enrichment analysis (GSEA) analysis also showed that the mechanism of ACAP1 in regulating ovarian cancer was related to a variety of immune-related pathways. In addition, we also revealed that the expression of ACAP1 was altered during cell differentiation and associated with cancer cell stemness markers. Our study highlights the clinical significance of ACAP1 in ovarian cancer and provides insight into the novel function of ACAP1 in regulation of tumor immune microenvironment and cancer cell stemness.
Collapse
Affiliation(s)
- Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinyi Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jing Zhang
- Department of Integrated Therapy, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
22
|
Outcomes in Advanced Stage Epithelial Ovarian, Fallopian Tubal, and Peritoneal Cancer after Primary Surgery and Adjuvant Chemotherapies: A Single-Institute Real-World Experience. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17103523. [PMID: 32443497 PMCID: PMC7277645 DOI: 10.3390/ijerph17103523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022]
Abstract
Debulking surgery followed by systemic chemotherapy—including three-weekly intravenous paclitaxel and carboplatin (GOG-158)—is the cornerstone for advanced epithelial ovarian, fallopian tubal, and peritoneal cancer (EOC) treatment. In this scenario, Federation of Gynecology and Obstetrics (FIGO) stage, cell types, completeness of surgery, lymph nodes (LN) status, adjuvant chemotherapy regimens, survival status, progression-free survival (PFS), and overall survival (OS) of 192 patients diagnosed as having stage IIIA1–IVB EOC over January 2008–December 2017 were analyzed retrospectively. Of them, 100 (52.1%) patients had been debulked optimally. Of all cases, 64.1% and 10.9% demonstrated serous and clear-cell carcinoma. Moreover, the FIGO stage, surgery completeness, and LN status affected recurrence/persistence and mortality (all p < 0.001). Clear cell carcinoma led to shorter survival than serous carcinoma (p = 0.002). Adjuvant chemotherapy regimens were divided into five main groups according to previous clinical trials. However, choice of chemotherapy failed to demonstrate significant differences in patient outcomes. Similar results were found in the sub-analysis of optimally debulked cases, except that intraperitoneal chemotherapy could reduce mortality risk when compared with GOG-158 (p = 0.042). Notably, retroperitoneal LN dissection in all cases or optimally debulked cases reduced risks of recurrence/persistence and mortality, and prolonged PFS and OS significantly (all p < 0.05). Without optimal debulking, LN dissection led to little improvement in outcomes. Various modified chemotherapy regimens did not prolong PFS and OS or reduce recurrence/persistence and mortality risks. LN dissection is strongly recommended to improve the completeness of surgery and patient outcome. Clear cell type has a poorer outcome than serous type, which requires more aggressive treatment and follow-up.
Collapse
|
23
|
Huang CY, Cheng M, Lee NR, Huang HY, Lee WL, Chang WH, Wang PH. Comparing Paclitaxel-Carboplatin with Paclitaxel-Cisplatin as the Front-Line Chemotherapy for Patients with FIGO IIIC Serous-Type Tubo-Ovarian Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17072213. [PMID: 32224896 PMCID: PMC7177627 DOI: 10.3390/ijerph17072213] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
The use of weekly chemotherapy for the treatment of patients with advanced-stage serous-type epithelial Tubo-ovarian cancer (ETOC), and primary peritoneal serous carcinoma (PPSC) is acceptable as the front-line postoperative chemotherapy after primary cytoreductive surgery (PCS). The main component of dose-dense chemotherapy is weekly paclitaxel (80 mg/m2), but it would be interesting to know what is the difference between combination of triweekly cisplatin (20 mg/m2) or triweekly carboplatin (carboplatin area under the curve 5-7 mg/mL per min [AUC 5-7]) in the dose-dense paclitaxel regimen. Therefore, we compared the outcomes of women with Gynecology and Obstetrics (FIGO) stage IIIC ETOC and PPSC treated with PCS and a subsequent combination of dose-dense weekly paclitaxel and triweekly cisplatin (paclitaxel–cisplatin) or triweekly carboplatin using AUC 5 (paclitaxel–carboplatin). Between January 2010 and December 2016, 40 women with International Federation of Gynecology and Obstetrics (FIGO) stage IIIC EOC, FTC, or PPSC were enrolled, including 18 treated with paclitaxel–cisplatin and the remaining 22 treated with paclitaxel–carboplatin. There were no statistically significant differences in disease characteristics of patients between two groups. Outcomes in paclitaxel–cisplatin group seemed to be little better than those in paclitaxel–carboplatin (median progression-free survival [PFS] 30 versus 25 months as well as median overall survival [OS] 58.5 versus 55.0 months); however, neither reached a statistically significant difference. In terms of adverse events (AEs), patients in paclitaxel–carboplatin group had more AEs, with a higher risk of neutropenia and grade 3/4 neutropenia, and the need for a longer period to complete the front-line chemotherapy, and the latter was associated with worse outcome for patients. We found that a period between the first-time chemotherapy to the last dose (6 cycles) of chemotherapy >21 weeks was associated with a worse prognosis in patients compared to that ≤21 weeks, with hazard ratio (HR) of 81.24 for PFS and 9.57 for OS. As predicted, suboptimal debulking surgery (>1 cm) also contributed to a worse outcome than optimal debulking surgery (≤1 cm) with HR of 14.38 for PFS and 11.83 for OS. Based on the aforementioned findings, both regimens were feasible and effective, but maximal efforts should be made to achieve optimal debulking surgery and following the on-schedule administration of dose-dense weekly paclitaxel plus triweekly platinum compounds. Randomized trials validating the findings are warranted.
Collapse
Affiliation(s)
- Chen-Yu Huang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; (C.-Y.H.); (M.C.)
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan;
| | - Min Cheng
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; (C.-Y.H.); (M.C.)
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan;
| | - Na-Rong Lee
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; (C.-Y.H.); (M.C.)
- Department of Nursing, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Hsin-Yi Huang
- Biostatics Task Force, Taipei Veterans General Hospital, Taipei 112, Taiwan;
| | - Wen-Ling Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Medicine, Cheng-Hsin General Hospital, Taipei 112, Taiwan
- Department of Nursing, Oriental Institute of Technology, New Taipei City 220, Taiwan
| | - Wen-Hsun Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; (C.-Y.H.); (M.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Nursing, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Correspondence: (W.-H.C.); (P.-H.W.); Tel.: +886-2-2875-7826 (W.-H.C.); +886-2-2875-7566 (P.-H.W.)
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; (C.-Y.H.); (M.C.)
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Medical Research, China Medical University Hospital, Taichung 440, Taiwan
- Female Cancer Foundation, Taipei 104, Taiwan
- Correspondence: (W.-H.C.); (P.-H.W.); Tel.: +886-2-2875-7826 (W.-H.C.); +886-2-2875-7566 (P.-H.W.)
| |
Collapse
|