1
|
Sjoerdsma JN, Bromley EK, Shin J, Hilliard T, Liu Y, Horgan C, Hwang G, Bektas M, Omstead D, Kiziltepe T, Stack MS, Bilgicer B. Combination non-targeted and sGRP78-targeted nanoparticle drug delivery outperforms either component to treat metastatic ovarian cancer. J Control Release 2024; 375:438-453. [PMID: 39271060 PMCID: PMC11486564 DOI: 10.1016/j.jconrel.2024.09.014] [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: 04/16/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Metastatic ovarian cancer (MOC) is highly deadly, due in part to the limited efficacy of standard-of-care chemotherapies to metastatic tumors and non-adherent cancer cells. Here, we demonstrated the effectiveness of a combination therapy of GRP78-targeted (TNPGRP78pep) and non-targeted (NP) nanoparticles to deliver a novel DM1-prodrug to MOC in a syngeneic mouse model. Cell surface-GRP78 is overexpressed in MOC, making GRP78 an optimal target for selective delivery of nanoparticles to MOC. The NP + TNPGRP78pep combination treatment reduced tumor burden by 15-fold, compared to untreated control. Increased T cell and macrophage levels in treated groups also suggested antitumor immune system involvement. The NP and TNPGRP78pep components functioned synergistically through two proposed mechanisms of action. The TNPGRP78pep targeted non-adherent cancer cells in the peritoneal cavity, preventing the formation of new solid tumors, while the NP passively targeted existing solid tumor sites, providing a sustained release of the drug to the tumor microenvironment.
Collapse
Affiliation(s)
- Jenna N Sjoerdsma
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily K Bromley
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jaeho Shin
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tyvette Hilliard
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Caitlin Horgan
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Gyoyeon Hwang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael Bektas
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David Omstead
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tanyel Kiziltepe
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - M Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Basar Bilgicer
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Berthiaume Institute for Precision Health, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Rare & Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
2
|
Geethadevi A, Ku Z, Tsaih SW, Parashar D, Kadamberi IP, Xiong W, Deng H, George J, Kumar S, Mittal S, Zhang N, Pradeep S, An Z, Chaluvally-Raghavan P. Blocking Oncostatin M receptor abrogates STAT3 mediated integrin signaling and overcomes chemoresistance in ovarian cancer. NPJ Precis Oncol 2024; 8:127. [PMID: 38839865 PMCID: PMC11153533 DOI: 10.1038/s41698-024-00593-y] [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: 01/07/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Chemotherapy such as cisplatin is widely used to treat ovarian cancer either before or after surgical debulking. However, cancer relapse due to chemotherapy resistance is a major challenge in the treatment of ovarian cancer. The underlying mechanisms related to chemotherapy resistance remain largely unclear. Therefore, identification of effective therapeutic strategies is urgently needed to overcome therapy resistance. Transcriptome-based analysis, in vitro studies and functional assays identified that cisplatin-resistant ovarian cancer cells express high levels of OSMR compared to cisplatin sensitive cells. Furthermore, OSMR expression associated with a module of integrin family genes and predominantly linked with integrin αV (ITGAV) and integrin β3 (ITGB3) for cisplatin resistance. Using ectopic expression and knockdown approaches, we proved that OSMR directly regulates ITGAV and ITGB3 gene expression through STAT3 activation. Notably, targeting OSMR using anti-OSMR human antibody inhibited the growth and metastasis of ovarian cancer cells and sensitized cisplatin treatment. Taken together, our results underscore the pivotal role of OSMR as a requirement for cisplatin resistance in ovarian cancer. Notably, OSMR fostered the expression of a distinct set of integrin genes, which in turn resulted into a crosstalk between OSMR and integrins for signaling activation that is critical for cisplatin resistance. Therefore, targeting OSMR emerges as a promising and viable strategy to reverse cisplatin-resistance in ovarian cancer.
Collapse
Affiliation(s)
- Anjali Geethadevi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ishaque P Kadamberi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Jasmine George
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sudhir Kumar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sonam Mittal
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA.
| | - Pradeep Chaluvally-Raghavan
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
3
|
Thomas DS, Sebastian A, Thomas V, Thomas A, Chandy RG, Peedicayil A. Diagnostic Accuracy of CA 19-9 in Ovarian Malignancy. INDIAN JOURNAL OF GYNECOLOGIC ONCOLOGY 2024; 22:73. [DOI: 10.1007/s40944-024-00829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/24/2024] [Accepted: 04/07/2024] [Indexed: 01/03/2025]
|
4
|
Kikuchi Y, Shimada H, Yamasaki F, Yamashita T, Araki K, Horimoto K, Yajima S, Yashiro M, Yokoi K, Cho H, Ehira T, Nakahara K, Yasuda H, Isobe K, Hayashida T, Hatakeyama S, Akakura K, Aoki D, Nomura H, Tada Y, Yoshimatsu Y, Miyachi H, Takebayashi C, Hanamura I, Takahashi H. Clinical practice guidelines for molecular tumor marker, 2nd edition review part 2. Int J Clin Oncol 2024; 29:512-534. [PMID: 38493447 DOI: 10.1007/s10147-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024]
Abstract
In recent years, rapid advancement in gene/protein analysis technology has resulted in target molecule identification that may be useful in cancer treatment. Therefore, "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" was published in Japan in September 2021. These guidelines were established to align the clinical usefulness of external diagnostic products with the evaluation criteria of the Pharmaceuticals and Medical Devices Agency. The guidelines were scoped for each tumor, and a clinical questionnaire was developed based on a serious clinical problem. This guideline was based on a careful review of the evidence obtained through a literature search, and recommendations were identified following the recommended grades of the Medical Information Network Distribution Services (Minds). Therefore, this guideline can be a tool for cancer treatment in clinical practice. We have already reported the review portion of "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" as Part 1. Here, we present the English version of each part of the Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Department of Surgery, Toho University, Tokyo, Japan.
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taku Yamashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Koji Araki
- Department of Otorhinolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Keigo Yokoi
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Takuya Ehira
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazunari Nakahara
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroshi Yasuda
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazutoshi Isobe
- Division of Respiratory Medicine, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | | | - Daisuke Aoki
- International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Yuji Tada
- Department of Pulmonology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center Research Institute, Tochigi, Japan
| | - Hayato Miyachi
- Faculty of Clinical Laboratory Sciences, Nitobe Bunka College, Tokyo, Japan
| | - Chiaki Takebayashi
- Division of Hematology and Oncology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | | |
Collapse
|
5
|
Zebic DS, Tjokrowidjaja A, Francis KE, Friedlander M, Gebski V, Lortholary A, Joly F, Hasenburg A, Mirza M, Denison U, Cecere SC, Ferrero A, Pujade-Lauraine E, Lee CK. Discordance between GCIG CA-125 progression and RECIST progression in the CALYPSO trial of patients with platinum-sensitive recurrent ovarian cancer. Br J Cancer 2024; 130:425-433. [PMID: 38097739 PMCID: PMC10844635 DOI: 10.1038/s41416-023-02528-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND CA-125 alone is widely used to diagnose progressive disease (PD) in platinum-sensitive recurrent ovarian cancer (PSROC) on chemotherapy. However, there are increasing concerns regarding its accuracy. We assessed concordance between progression defined by CA-125 and RECIST using data from the CALYPSO trial. METHODS We computed concordance rates for PD by CA-125 and RECIST to determine the positive (PPV) and negative predictive values (NPV). RESULTS Of 769 (79%) evaluable participants, 387 had CA-125 PD, where only 276 had concordant RECIST PD (PPV 71%, 95% CI 67-76%). For 382 without CA-125 PD, 255 had RECIST PD but 127 did not (NPV 33%, 95% CI 29-38). There were significant differences in NPV according to baseline CA-125 (≤100 vs >100: 42% vs 25%, P < 0.001); non-measurable vs measurable disease (51% vs 26%, P < 0.001); and platinum-free-interval (>12 vs 6-12 months: 41% vs 14%, P < 0.001). We observed falling CA-125 levels in 78% of patients with RECIST PD and CA-125 non-PD. CONCLUSION Approximately 2 in 3 women with PSROC have RECIST PD but not CA-125 PD by GCIG criteria. Monitoring CA-125 levels alone is not reliable for detecting PD. Further research is required to investigate the survival impact of local therapy in radiological detected early asymptomatic PD.
Collapse
Affiliation(s)
- Danka Sinikovic Zebic
- National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, Sydney, NSW, 2050, Australia.
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, 2217, Australia.
| | - Angelina Tjokrowidjaja
- National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, Sydney, NSW, 2050, Australia
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Katherine Elizabeth Francis
- National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, Sydney, NSW, 2050, Australia
- Department of Medical Oncology, South East Regional Hospital, Bega, NSW, 2550, Australia
| | - Michael Friedlander
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Val Gebski
- National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, Sydney, NSW, 2050, Australia
| | | | - Florence Joly
- Centre François Baclesse, Caen and GINECO, Caen, France
| | - Annette Hasenburg
- Department of Gynecology and Obstetrics, University Medical Center, Mainz and AGO, Mainz, Germany
| | - Mansoor Mirza
- Rigshospitalet-Copenhagen University Hospital, Copenhagen and NSGO, Copenhagen, Denmark
| | - Ursula Denison
- Institute for gynaecological oncology und senology - Karl Landsteiner, Vienna and AGO Austria, Vienna, Austria
| | - Sabrina Chiara Cecere
- Oncologia Clinica Sperimentale Uro-Ginecologica, Istituto Nazionale Tumori - IRCCS- Fondazione G.Pascale, Napoli and MITO Italia, Napoli, Italy
| | - Annamaria Ferrero
- Academic Division Gynaecology, Mauriziano Hospital, University of Torino, and MaNGO, Torino, Italy
| | | | - Chee Khoon Lee
- National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, Sydney, NSW, 2050, Australia
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, 2217, Australia
| |
Collapse
|
6
|
Brzezinska B, Mysona DP, Richardson KP, Rungruang B, Hopkins D, Bearden G, Higgins R, Johnson M, Satter KB, McIndoe R, Ghamande S, Purohit S. High serum levels of inflammatory markers are associated with early recurrence in patients with high-grade serous ovarian cancer after platinum therapy. Gynecol Oncol 2023; 179:1-8. [PMID: 37862814 DOI: 10.1016/j.ygyno.2023.10.009] [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: 09/14/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
OBJECTIVE To determine if inflammatory biomarkers can predict the long-term outcome of platinum therapy in patients with high-grade serous ovarian cancer. METHODS Women diagnosed with high-grade serous epithelial ovarian cancer (n = 70) at a single institution were enrolled in a prospective serum collection study between 2005 and 2020. Seventeen markers of inflammation and oxidative stress were measured in serum samples on a chemistry analyzer. Association was tested for serum levels with progression-free survival (PFS), time to recurrence (TTR), overall survival (OS), and time to death (TTD) using Cox proportional hazards and Kaplan-Meier curves. Patient survival was censored at 10 years. RESULTS Higher serum levels of LDH were associated with worse PFS (HR 2.57, p = 0.028). High serum levels of BAP (HR 0.38, p = 0.025), GSP (HR 0.40, p = 0.040), HDL-c (HR 0.27, p = 0.002), and MG (HR 0.36, p = 0.017) were associated with improved PFS. Higher expression of LDH was associated with worse OS (HR 2.16, p = 0.023). Higher levels of CK.nac (HR 0.39, p = 0.033) and HDL-c (HR 0.35, p = 0.029) were associated with improved OS. Similar outcomes were found with TTR and TTD analyses. CONCLUSION General inflammatory biomarkers may serve as a guide for prognosis and treatment benefit. Future studies needed to further define their role in predicting prognosis or how these markers may affect response to therapy.
Collapse
Affiliation(s)
- Bogna Brzezinska
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David P Mysona
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Katherine P Richardson
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Bunja Rungruang
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Diane Hopkins
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gregory Bearden
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Robert Higgins
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Marian Johnson
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Khaled Bin Satter
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Richard McIndoe
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sharad Ghamande
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sharad Purohit
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Raheem KY, Ibukunoluwa FP, Olorundare SA, Nandwa JO, Abayomi MA, Uchechukwu EJ, Adewunmi M, Blessing KZ, Anthony MM, Gbadebo MI, Daniel FT. Therapeutic capability of selected medicinal plants' bioactive constituents against the mutant ovarian TP53 gene; a computational approach. ADVANCES IN BIOMARKER SCIENCES AND TECHNOLOGY 2023. [DOI: 10.1016/j.abst.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
8
|
Zhang J, Li W, Xiao J, Hui J, Li Y. Prognostic significance of carbohydrate antigen 125 in stage D heart failure. BMC Cardiovasc Disord 2023; 23:108. [PMID: 36841766 PMCID: PMC9960178 DOI: 10.1186/s12872-023-03139-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND The predictive value of carbohydrate antigen 125 (CA125) has not been examined in stage D heart failure (HF) patients, regardless of left ventricular ejection fraction (LVEF). We sought to quantify the prognostic usefulness in predicting death and HF readmission in this cohort. METHODS According to CA125 levels above and below the median (65.7 U/ml), 176 stage D HF patients including more than half (50.6%) had LVEF > 40% were divided into 2 groups. RESULTS A total of 106 (60.2%) deaths and 102 deaths due to the cardiovascular disease were identified. All-cause death/HF readmission and MACE occurred in 157 patients (89.2%) during 18 months (16-20) of follow-up. By the Kaplan-Meier method, subjects with CA125 ≥ 65.7 U/ml exhibited higher 1-year mortality rate (59.3% vs. 31.0%, P < 0.001) and 1-year death/HF rehospitalization rate (94.2% vs. 80.6%, P < 0.001). In univariate Cox analysis, CA125 (categorized) was a significant prognostic factor for all-cause death, cardiovascular mortality, death/HF readmission and MACE. Based on multivariate Cox analysis, elevated CA125 was still significant for all-cause death, cardiovascular mortality, death/HF readmission and MACE. CONCLUSIONS In stage D HF patients, elevated CA125 levels were highly predictive of all-cause death, cardiovascular mortality, all-cause death/HF readmission and MACE, which can be used for better risk stratification.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Cardiology, Wujin Hospital Affiliated With Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, No. 2, Yongning Road, Changzhou, Jiangsu Province, China.
| | - Wenhua Li
- Department of Cardiology, Wujin Hospital Affiliated With Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, No. 2, Yongning Road, Changzhou, Jiangsu Province, China
| | - Jianqiang Xiao
- Department of Cardiology, Wujin Hospital Affiliated With Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, No. 2, Yongning Road, Changzhou, Jiangsu Province, China
| | - Jie Hui
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Li
- School of Math and Statistics, Nanjing Audit University, Nanjing, China
| |
Collapse
|
9
|
DelaCourt A, Mehta A. Beyond glyco-proteomics-Understanding the role of genetics in cancer biomarkers. Adv Cancer Res 2023; 157:57-81. [PMID: 36725113 DOI: 10.1016/bs.acr.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The development of robust cancer biomarkers is the most effective way to improve overall survival, as early detection and treatment leads to significantly better clinical outcomes. Many of the cancer biomarkers that have been identified and are clinically utilized are glycoproteins, oftentimes a specific glycoform. Aberrant glycosylation is a common theme in cancer, with dysregulated glycosylation driving tumor initiation and metastasis, and abnormal glycosylation can be detection both on the tissue surface and in serum. However, most cancer types are heterogeneous in regard to tumor genomics, and this heterogeneity extends to cancer glycomics. This limits the sensitivity of standalone glycan-based biomarkers, which has slowed their implementation clinically. However, if targeted biomarker development can take into account genomic tumor information, the development of complementary biomarkers that target unique cancer subgroups can be accomplished. This idea suggests the need for algorithm-based cancer biomarkers, which can utilize multiple biomarkers along with relevant demographic information. This concept has already been established in the detection of hepatocellular carcinoma with the GALAD score, and an algorithm-based approach would likely be effective in improving biomarker sensitivity for additional cancer types. In order to increase cancer diagnostic biomarker sensitivity, there must be more targeted biomarker development that considers tumor genomic, proteomic, metabolomic, and clinical data while identifying tumor biomarkers.
Collapse
Affiliation(s)
- Andrew DelaCourt
- Department of Cell & Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Anand Mehta
- Department of Cell & Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
10
|
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
|
11
|
Kennedy F, Shearsmith L, Holmes M, Velikova G. 'It made me feel part of the team, having my homework to do' - women and specialist nurse experiences of remote follow-up after ovarian cancer treatment: a qualitative interview study. Support Care Cancer 2022; 31:2. [PMID: 36512093 PMCID: PMC9745768 DOI: 10.1007/s00520-022-07470-z] [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: 07/12/2022] [Accepted: 11/07/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE Ovarian cancer patients require monitoring for relapse post-treatment, and alternative follow-up pathways are increasing, which require in-depth exploration to ensure acceptability and inform implementation. This study aimed to explore women and specialist nurses' experiences of participating in a feasibility study of an electronic patient-reported outcome (ePRO) follow-up pathway after ovarian cancer treatment. METHODS The feasibility study incorporated an ePRO questionnaire, blood test and telephone consultation with a specialist nurse, instead of face-to-face hospital visits. All women and the nurses involved were invited to take part in nested semi-structured interviews. Interviews were recorded and transcripts analysed using framework analysis. RESULTS Twenty interviews were conducted (16 out of 24 women who took part in the feasibility study and all 4 nurses). Four themes were identified: (1) readiness and motivators, (2) practicalities and logistics, (3) personal impact and (4) future role. An overarching theme highlighted how women strived to seek reassurance and gain confidence. Most women and nurses were positive about the ePRO pathway and would happily continue using it. CONCLUSION This work provides invaluable insight into the experiences of women on remote ePRO follow-up post-treatment. Important logistic and implementation issues were identified, which should inform future large-scale work to introduce and evaluate remote ePRO methods in cancer follow-up. This work highlights the key factors influencing women's readiness and acceptability of an ePRO pathway, and how services should be carefully designed to ensure patients feel reassured and confident post-treatment. Furthermore, it highlights that flexibility and patient preference should be considered in remote service delivery. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT02847715 (first registered 19 May 2016).
Collapse
Affiliation(s)
- Fiona Kennedy
- Section of Patient Centred Outcomes Research, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK.
| | - Leanne Shearsmith
- Section of Patient Centred Outcomes Research, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Marie Holmes
- Section of Patient Centred Outcomes Research, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Galina Velikova
- Section of Patient Centred Outcomes Research, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
12
|
Chen X, Wang L, Liu S, Luo X, Wang K, He Q. Cisplatin-loaded metal–phenolic network with photothermal-triggered ROS generation for chemo-photothermal therapy of cancer. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00149-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
AbstractDeveloping multifunctional composites has received widespread attention for cancer treatment. Herein, a metal–phenolic network (MPN)-based composite loading with chemotherapy agents (TAFP) exhibits high anti-tumor therapeutic efficacy via photothermal therapy (PTT), chemo-dynamic therapy (CDT), and chemotherapy. The nanocomposite was formed by mixing the chemotherapeutic drugs (cisplatin, DDP) into the tannic acid (TA) and Fe3+ network (TAFe) to integrate the synergistic effect of PTT, CDT, and chemotherapy. Due to the acidic tumor microenvironment, the active substances could be released with the degradation of the metal–phenolic network, and the released DDP would induce the chemotherapy. More importantly, the released TA under the acidic environment could increase iron bioavailability by converting Fe3+ to Fe2+, which converts hydrogen peroxide (H2O2) to highly toxic hydroxyl radical via the Fenton reaction. Meanwhile, the heat generated from TAFP after near-infrared (NIR) laser irradiation could enhance the therapeutic effect of CDT and chemotherapy. Furthermore, the composite exhibited unique anticancer efficacy in vivo with low toxicity. Collectively, this work may facilitate the development of metal–phenolic network-based photothermal agents for clinic anti-tumor applications.
Collapse
|
13
|
TÜRKCAN C, AKGÖL S. Polymeric nanoparticle-based electrochemical sensor for the detection of CA 125. Turk J Chem 2022; 47:137-147. [PMID: 37720868 PMCID: PMC10504010 DOI: 10.55730/1300-0527.3524] [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: 05/17/2022] [Revised: 02/20/2023] [Accepted: 11/02/2022] [Indexed: 02/25/2023] Open
Abstract
In this paper, Cys-graft-p(HEMA) nanomaterials and a new electrochemical method were developed for determination of CA 125. Cys-graft-p(HEMA) nanomaterials were synthesized with emulsion polymerization method and modified with grafting procedure. It was determined that Cys-graft-p(HEMA) nanomaterials had 50 nm dimension and spherical morphology, and per gram polymeric material contained 0.011 mmol L-cysteine. Electrode surface was prepared step by step for electrochemical analysis with optimization process. Linear determination range was determined as 5-400 U/mL (R= 0.9935). Detection limit (LOD) was calculated as 1.87 U/mL, and quantification limit (LOQ) was determined as 5.62 U/mL. The fabricated sensor system showed good repeatability, accuracy, reality, and storage stability. According to the results obtained, Cys-graft p(HEMA) nanomaterials that is used for the first time in biosensor has the potential to find use in the sector with rapid determination time (10 min), extensive determination range, accuracy of methods. Novelties of this study are rapid analysis, determination range, appropriate of prototype device development, and developing new designed material. Developed material and method can be used in the preliminary diagnosis of the disease and combined with a prototype device that can allow the follow-up of the treatment process in diagnosed patients.
Collapse
Affiliation(s)
- Ceren TÜRKCAN
- Departmet of Biomedical Engineering, Faculty of Engineering and Architecture, İstanbul Arel University, İstanbul,
Turkey
| | - Sinan AKGÖL
- Department of Biochemistry, Faculty of Science, Ege University, İzmir,
Turkey
| |
Collapse
|
14
|
Janke MJ, Santiago S, Straubhar AM, Uppal S. The utility of physical examination in ovarian cancer recurrence detection: a retrospective analysis informing virtual surveillance care. Int J Gynecol Cancer 2022; 32:913-917. [PMID: 35675968 DOI: 10.1136/ijgc-2022-003506] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Given the recent rapid increase in telemedicine in the setting of the COVID-19 pandemic, we sought to investigate the utility of symptom review, CA125, and physical examination in the detection of ovarian cancer recurrence to determine the role of virtual surveillance care in the COVID-19 era. METHODS This retrospective cohort study included patients diagnosed with ovarian cancer between 2013 and 2020 who achieved remission after primary treatment and then had recurrence while in a routine surveillance program. Modalities that detected recurrence including symptoms, CA125, physical examination, or 'other,' which was denoted if imaging was obtained for reasons other than suspected recurrence and recurrence was incidentally identified, were recorded. Descriptive statistics were performed to summarize the cohort. RESULTS One hundred and nine patients met inclusion criteria. At time of recurrence, elevated CA125 was present in 97 (89.0%) patients, symptoms in 41 (37.6%), and abnormal physical exam findings in 27 (24.8%). Recurrence was incidentally found with imaging obtained for reasons other than suspicion of recurrence in six (5.5%) patients. Recurrence was suspected based on multiple modalities in 46 (42.2%) patients. Elevated CA125, symptoms, or both were present in 102 (93.6%) patients. Of patients with abnormal physical exam findings, 26 (96.3%) also had elevated CA125 or symptoms present. Recurrence was suspected based on physical exam findings alone in one (0.9%) patient. CONCLUSIONS Over 90% of ovarian cancer recurrences were detected by rising CA125, symptoms, or both. Only one patient had recurrence detected by physical examination alone. Given that review of symptoms and CA125 can be conducted virtually, virtual visits may offer a reasonable alternative to in-person visits for ovarian cancer surveillance for patients who have pre-treatment elevated CA125.
Collapse
Affiliation(s)
- Monica J Janke
- Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Santiago
- Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alli M Straubhar
- Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Shitanshu Uppal
- Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Kennedy F, Shearsmith L, Holmes M, Rogers Z, Carter R, Hofmann U, Velikova G. Electronic patient-reported monitoring of symptoms during follow-up of ovarian cancer patients: a feasibility study. BMC Cancer 2022; 22:726. [PMID: 35780095 PMCID: PMC9250717 DOI: 10.1186/s12885-022-09817-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background Ovarian cancer patients require monitoring for relapse. Innovative follow-up methods are increasingly being explored. An electronic patient-reported outcome (ePRO) follow-up pathway was developed for women treated for ovarian cancer. This feasibility study explored patient acceptability and compliance. Methods A single-arm non-blinded prospective feasibility study was undertaken at two hospitals. Participants were women who had completed treatment for ovarian cancer whose clinician was happy for them to be monitored remotely. Automated 3-monthly reminders were sent to participants to complete an ePRO questionnaire and obtain blood tests. Participants were reviewed over the phone by their clinical nurse specialist instead of attending clinic-based follow-up. The primary outcome was compliance (expected ePRO completions/blood tests) across the 12-month study period. Secondary outcomes were recruitment, attrition, resource use, symptom severity/alerts and patient acceptability. Results Twenty-four women consented (50% consent rate), and 13 remained on study at 12 months. Seven women relapsed, 3 chose to withdraw, and 1 withdrew for other clinical reasons. ePRO compliance was high and consistent at 75-82%, although the two hospitals differed. Adherence to the clinical protocol was evident for blood tests and contacts with staff (fewer visits, more phonecalls compared to an earlier audit). End-of-study feedback indicated high patient satisfaction. Conclusions Remote ePRO follow-up for ovarian cancer is feasible and acceptable to patients who are able and willing to participate. However, the low recruitment rate (ineligible + declined) indicate it is not suitable/acceptable to all patients immediately post-treatment. Further large-scale research and implementation work is required, especially in a post-COVID era. Trial registration ClinicalTrials.gov ID: NCT02847715 (first registered 19/05/2016). Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09817-5.
Collapse
Affiliation(s)
- Fiona Kennedy
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK.
| | - Leanne Shearsmith
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Marie Holmes
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Zoe Rogers
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Rob Carter
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Uschi Hofmann
- Calderdale & Huddersfield NHS Foundation Trust, Huddersfield Royal Infirmary, Acre St, Lindley, Huddersfield, HD3 3EA, UK
| | - Galina Velikova
- Section of Patient Centred Outcomes Research, Patient Reported Outcomes Group, Leeds Institute of Medical Research at St James's, University of Leeds, Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
16
|
Recent Advances in Ovarian Cancer: Therapeutic Strategies, Potential Biomarkers, and Technological Improvements. Cells 2022; 11:cells11040650. [PMID: 35203301 PMCID: PMC8870715 DOI: 10.3390/cells11040650] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Aggressive and recurrent gynecological cancers are associated with worse prognosis and a lack of effective therapeutic response. Ovarian cancer (OC) patients are often diagnosed in advanced stages, when drug resistance, angiogenesis, relapse, and metastasis impact survival outcomes. Currently, surgical debulking, radiotherapy, and/or chemotherapy remain the mainstream treatment modalities; however, patients suffer unwanted side effects and drug resistance in the absence of targeted therapies. Hence, it is urgent to decipher the complex disease biology and identify potential biomarkers, which could greatly contribute to making an early diagnosis or predicting the response to specific therapies. This review aims to critically discuss the current therapeutic strategies for OC, novel drug-delivery systems, and potential biomarkers in the context of genetics and molecular research. It emphasizes how the understanding of disease biology is related to the advancement of technology, enabling the exploration of novel biomarkers that may be able to provide more accurate diagnosis and prognosis, which would effectively translate into targeted therapies, ultimately improving patients’ overall survival and quality of life.
Collapse
|
17
|
Kennedy F, Shearsmith L, Holmes M, Peacock R, Lindner OC, Megson M, Velikova G. 'We do need to keep some human touch'-Patient and clinician experiences of ovarian cancer follow-up and the potential for an electronic patient-reported outcome pathway: A qualitative interview study. Eur J Cancer Care (Engl) 2022; 31:e13557. [PMID: 35146821 PMCID: PMC9287040 DOI: 10.1111/ecc.13557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/30/2021] [Accepted: 01/26/2022] [Indexed: 12/29/2022]
Abstract
Objective This study aimed to explore experiences of follow‐up after treatment and views on an electronic patient‐reported outcome (ePRO) pathway among ovarian cancer patients and clinicians. Methods Semi‐structured qualitative interviews were conducted with clinicians and patients previously treated for ovarian cancer. Interviews explored experiences of the current follow‐up pathway, patients' needs and views on an ePRO pathway enabling patients to report symptoms online rather than attend clinic‐based appointments. Transcripts were analysed using framework analysis. Results Sixteen patients and 10 clinicians participated from four hospitals in England. Four key themes were identified: transition into follow‐up, key features of effective follow‐up, issues in follow‐up and views of ePRO. Both patients and clinicians saw benefits of an ePRO pathway alongside continued access to specialist support and discussed various practicalities (e.g., frequency, introduction and communication). Technology concerns and feelings of abandonment were highlighted as barriers. The proposed impact on clinical and individual patient outcomes was discussed. Conclusion Patient and clinician views on follow‐up and an ePRO pathway informed key recommendations on the development/introduction of ePRO follow‐up. Technology use in healthcare will continue to grow and may offer solutions to facilitate responsive and tailored care. Further research should explore the safety, experiences and acceptability of ePRO follow‐up.
Collapse
Affiliation(s)
- Fiona Kennedy
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK
| | - Leanne Shearsmith
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK.,Academic Unit of Palliative Care, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Marie Holmes
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK
| | - Rosemary Peacock
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK
| | - Oana C Lindner
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK
| | - Molly Megson
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK.,Academic Unit of Palliative Care, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Galina Velikova
- Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, Leeds, UK
| |
Collapse
|
18
|
Chung YS, Kim Y, Kim HS, Lee JY, Kang WJ, Kim S, Kim SW. Prognostic value of complete metabolic response on 18F-FDG-PET/CT after three cycles of neoadjuvant chemotherapy in advanced high-grade serous ovarian cancer. J Gynecol Oncol 2022; 33:e28. [PMID: 35128858 PMCID: PMC9024185 DOI: 10.3802/jgo.2022.33.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/06/2021] [Accepted: 12/26/2021] [Indexed: 11/30/2022] Open
Abstract
Objective We investigated the prognostic value of complete metabolic response (CMR) on 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) after 3 cycles of neoadjuvant chemotherapy (NAC) in advanced high-grade serous ovarian cancer (HGSC). Methods PET/CT at baseline and after 3 cycles of NAC were performed; peak standardized uptakes were measured. PET parameters were compared with NAC parameter: cancer antigen-125 (CA-125) normalization before interval debulking surgery (IDS) and chemotherapy response score (CRS) to predict platinum-sensitivity. Kaplan-Meier analysis was used to determine correlations between PET parameters and survival. Prognostic factors were obtained by multivariate Cox regression analysis. Results Between 2007 and 2020, 102 patients were recruited: 19 (18.6%) were designated as CMR group and 83 (81.4%) as non-CMR group. CMR after 3 cycles of NAC showed the highest accuracy in predicting platinum-sensitivity (area under the curve [AUC]=0.729; 95% confidence interval [CI]=0.552–0.823; p=0.017), compared with CA-125 normalization before IDS (AUC=0.626; 95% CI=0.542–0.758; p=0.010) and CRS (AUC=0.613; 95% CI=0.490–0.735; p=0.080). CMR demonstrated better prognosis than non-CMR in progression-free survival (PFS) (median PFS, 23.9 months vs. 16.4 months; p=0.021) and overall survival (OS) (median OS, not reached vs. 69.7 months; p=0.025). In multivariate analysis, CMR was associated with a lower risk of recurrence (adjusted hazard ratio [aHR]=0.50; 95% CI=0.27–0.92; p=0.027) and death (aHR=0.23; 95% CI=0.05–0.99; p=0.048). Conclusion CMR after 3 cycles of NAC can be a prognostic factor for both recurrence and death in advanced HGSC. • CMR after NAC can be used to determine the extent of surgery. • CMR after NAC provides higher accuracy for predicting platinum-sensitivity, compared with cancer antigen-125 normalization before IDS. • CMR after NAC can be useful for stratifying prognosis in advanced HGSC.
Collapse
Affiliation(s)
- Young Shin Chung
- Department of Obstetrics and Gynecology, Institution of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yup Kim
- Department of Obstetrics and Gynecology, Institution of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institution of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Institution of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Wun Kim
- Department of Obstetrics and Gynecology, Institution of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Deng L, Wang D, Chen S, Hu W, Zhang R. Epiphycan Predicts Poor Outcomes and Promotes Metastasis in Ovarian Cancer. Front Oncol 2021; 11:653782. [PMID: 34888227 PMCID: PMC8650094 DOI: 10.3389/fonc.2021.653782] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
The small leucine-rich proteoglycan (SLRP) family is widely expressed in extracellular matrix and aggravates tumor progression. However, epiphycan (EPYC), as a member of the SLRPs family, its biological function in cancer has not been confirmed. Thus, we aimed to clarify the role of EPYC in progression of ovarian cancer (OC), and further analyze the molecular mechanisms implicated in tumorigenesis. Here, we analyzed the differential expression genes of GSE38734, including 4 matched primary OC and metastatic tissues. We obtained OC RNAseqs data from the Cancer Genome Atlas (TCGA) and analyzed the correlation between EPYC expression and OC staging, pathological grading, etc. The expression of EPYC in OC and normal ovarian tissues was compared in Oncomine website. We used siRNAs to interfere the expression of EPYC in ovarian cancer cell line SKOV3. Scratch test, transwell-matrigel chamber, CCK8 assay were used to detect the changes of SKOV3 migration, invasion and proliferation ability after EPYC was interfered. We used R software to make GO and KEGG analysis of related genes of EPYC. We used the Hitpredict website to predict interacting proteins. The results showed that the expression of EPYC in metastatic ovarian cancer was higher than primary ovarian cancer, and that in primary cancer was higher than normal ovaries. After siRNA interferes with EPYC expression, the migration, invasion and proliferation of SKOV3 cells were weakened. EPYC mainly played a role in ECM organization, and involved in PI3K/Akt, focal adhesion signaling pathways. EPYC might interact with PLCG2 and CRK, and be involved in signal transduction.
Collapse
Affiliation(s)
- Lu Deng
- Department of Gynaecology, The Hospital of Obstetrics & Gynaecology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Dandan Wang
- Department of Gynecology and Obstetrics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Shouzhen Chen
- Department of Gynaecology, The Hospital of Obstetrics & Gynaecology, Fudan University, Shanghai, China
| | - Weiguo Hu
- Department of Gynaecology, The Hospital of Obstetrics & Gynaecology, Fudan University, Shanghai, China
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Bio-Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Openshaw MR, McVeigh TP. Non-invasive Technology Advances in Cancer-A Review of the Advances in the Liquid Biopsy for Endometrial and Ovarian Cancers. Front Digit Health 2021; 2:573010. [PMID: 34713045 PMCID: PMC8521848 DOI: 10.3389/fdgth.2020.573010] [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: 06/15/2020] [Accepted: 11/19/2020] [Indexed: 01/02/2023] Open
Abstract
Improving cancer survival rates globally requires improvements in disease detection and monitoring, with the aim of improving early diagnosis and prediction of disease relapse. Traditional means of detecting and monitoring cancers rely largely on imaging and, where possible, blood-based protein biomarkers, many of which are non-specific. Treatments are being improved by identification of inherited and acquired genomic aberrations in tumors, some of which can be targeted by newly developed therapeutic interventions. Treatment of gynecological malignancy is progressively moving toward personalized therapy, as exemplified by application of PARP-inhibition for patients with BRCA-deficient tubo-ovarian cancers, or checkpoint inhibition in patients with mismatch repair-deficient disease. However, the more recent discovery of a group of biomarkers described under the umbrella term of “liquid biopsy” promises significant improvement in our ability to detect and monitor cancers. The term “liquid biopsy” is used to describe an array of tumor-derived material found in blood plasma and other bodily fluids such as ascites, pleural fluid, saliva, and urine. It includes circulating tumors cells (CTCs), circulating nucleic acids including DNA, messenger RNA and micro RNAs, and extracellular vesicles (EVs). In this review, we discuss recent advancements in liquid biopsy for biomarker detection to help in diagnosis, prognosis, and planning of treatment of ovarian and endometrial cancer.
Collapse
Affiliation(s)
- Mark R Openshaw
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Terri P McVeigh
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
21
|
Targeted design of green carbon dot-CA-125 aptamer conjugate for the fluorescence imaging of ovarian cancer cell. Cell Biochem Biophys 2021; 80:75-88. [PMID: 34716880 DOI: 10.1007/s12013-021-01034-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
Aptamer-Carbon Dot (CD) bioconjugation is an attractive target-tracking strategy in detecting cell surface antigens. This study describes an effective imaging paradigm for CA-125 antigen imaging. Our experience encompasses green CD synthesis and characterization, CD-capture probe conjugation through covalent bonding, the hybridization linkage of CD-probe to aptamer and their coupling confirmation, and fluorescent targeted imaging of ovarian cancer cells. As a result, the synthesized CDs from lemon extract by hydrothermal reaction show average size of 2 nm with maximum fluorescence intensity at excitation/emission 360/450 nm. CD-probe construction was provided by functional group interactions of CD and probe via EDC/NHS chemistry. The linkage of CD-probe to aptamer was conducted by Watson-Crick nucleotide pairing. The assessment of CD-probe and CD-probe-aptamer fabrication was validated by the increase in surface roughness through AFM analysis, the diminish of fluorescence intensity of CD after bioconjugation, and particle size growth of the construct. Conjugates with negligible cytotoxicity, appropriate zeta potential, and good aptamer release were applied in cellular imaging. This targeted diagnosis method was employed the four reported DNA aptamers toward fluorescence intensity. The DOV-3 aptamer showed more qualified detection over other aptamer conjugates during fluorescent microscopy analysis. In conclusion, the CD-probe-aptamer conjugate applications as toxic-free method can open new horizons in fluorescent nano-imaging in the field of targeted cancer cell diagnosis.
Collapse
|
22
|
Rao S, Smith DA, Guler E, Kikano EG, Rajdev MA, Yoest JM, Ramaiya NH, Tirumani SH. Past, Present, and Future of Serum Tumor Markers in Management of Ovarian Cancer: A Guide for the Radiologist. Radiographics 2021; 41:1839-1856. [PMID: 34597221 DOI: 10.1148/rg.2021210005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability to accurately detect early ovarian cancer and subsequently monitor treatment response is essential to improving survival for patients with ovarian malignancies. Several serum tumor markers (STMs)-including cancer antigen 125 (CA-125), human epididymis protein 4 (HE4), cancer antigen 19-9 (CA 19-9), and carcinoembryonic antigen (CEA)-have been used as a noninvasive method of identifying ovarian cancer in conjunction with imaging. Although current guidelines do not recommend use of STMs as screening tools for ovarian cancer, these markers have clinical utility in both diagnosis and surveillance for women with ovarian cancer. CA-125 is the most commonly used STM; its level may be elevated in several types of ovarian cancer, including epithelial cell tumors, carcinosarcoma, teratomas, and secondary ovarian malignancies. An elevated level of CA 19-9 is associated with clear cell tumors, teratomas, and secondary malignancies. CEA is most commonly associated with mucinous ovarian cancers. Finally, HE4 is being increasingly used to identify certain subtypes of epithelial ovarian cancers, particularly serous and endometrioid tumors. Diagnosis of ovarian cancers relies on a combination of CA-125 levels and US findings, which include a large adnexal mass or high-risk features, including septa and increased vascularity. CT is preferred for staging and is used along with PET and STM monitoring for surveillance. Increasingly, MRI is being used to characterize ovarian lesions that are indeterminate at US or CT. The future of STM testing involves development of "liquid biopsies," in which plasma samples are analyzed for evidence of tumors, including circulating tumor DNA or tumor cells and tumor micro-RNA. When combined with traditional imaging techniques, liquid biopsies may lead to earlier diagnosis and improved survival. An invited commentary by Shinagare is available online. ©RSNA, 2021.
Collapse
Affiliation(s)
- Sanjay Rao
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Daniel A Smith
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Ezgi Guler
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Elias G Kikano
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Maharshi A Rajdev
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Jennifer M Yoest
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Nikhil H Ramaiya
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| | - Sree Harsha Tirumani
- From the Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, B114, Cleveland, OH 44106-4915 (S.R., D.A.S., E.G., E.G.K., M.A.R., N.H.R., S.H.T.); and Department of Pathology, Case Western Reserve University, Cleveland, Ohio (J.M.Y.)
| |
Collapse
|
23
|
Mallet E, Angeles MA, Cabarrou B, Chardin D, Viau P, Frigenza M, Navarro AS, Ducassou A, Betrian S, Martínez-Gómez C, Tanguy Le Gac Y, Chantalat E, Motton S, Ferron G, Barranger E, Gabiache E, Martinez A. Performance of Multiparametric Functional Imaging to Assess Peritoneal Tumor Burden in Ovarian Cancer. Clin Nucl Med 2021; 46:797-806. [PMID: 34238796 DOI: 10.1097/rlu.0000000000003785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of the study was to evaluate the clinical utility of pretreatment 18F-FDG PET/CT with quantitative evaluation of peritoneal metabolic cartography in relation to staging laparoscopy for ovarian carcinomatosis. PATIENTS AND METHODS A retrospective review of prospectively collected data from 84 patients with FIGO (International Federation of Gynecology and Obstetrics) stage IIIC to IV ovarian cancer was carried out. All patients had a double-blinded 18F-FDG PET/CT review. Discriminant capacity of metabolic parameters to identify peritoneal carcinomatosis in the 13 abdominal regions according to the peritoneal cancer index was estimated with area under the receiver operating characteristic curve (AUC). RESULTS The metabolic parameter showing the best trade-off between sensitivity and specificity to predict peritoneal extension compared with peritoneal cancer index score was the metabolic tumor volume (MTV), with a Spearman ρ equal to 0.380 (P < 0.001). The AUC of MTV to diagnose peritoneal involvement in the upper abdomen (regions 1, 2, and 3) ranged from 0.740 to 0.765. MTV AUC values were lower in the small bowel regions (9-12), ranging from 0.591 to 0.681, and decreased to 0.487 in the pelvic region 6. 18F-FDG PET/CT also improved the detection of extra-abdominal disease, upstaging 35 patients (41.6%) from stage IIIC to IV compared with CT alone and leading to treatment modification in more than one third of patients. CONCLUSIONS 18F-FDG PET/CT metrics are highly accurate to reflect peritoneal tumor burden, with variable diagnostic value depending on the anatomic region. MTV is the most representative metabolic parameter to assess peritoneal tumor extension.
Collapse
Affiliation(s)
- Estelle Mallet
- From the Department of Surgical Oncology, Centre Antoine Lacassagne, Nice
| | | | - Bastien Cabarrou
- Biostatistics, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse
| | - David Chardin
- Department of Nuclear Medicine, Centre Antoine Lacassagne
| | - Philippe Viau
- Department of Nuclear Medicine, Centre Hospitalier Universitaire de Nice
| | - Mélanie Frigenza
- Department of Gynecological Surgery, Centre Hospitalier Universitaire de Nice, Nice
| | | | | | - Sarah Betrian
- Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse
| | | | - Yann Tanguy Le Gac
- Department of Gynecology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole
| | - Elodie Chantalat
- Department of Gynecology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole
| | - Stéphanie Motton
- Department of Gynecology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole
| | | | - Emanuel Barranger
- From the Department of Surgical Oncology, Centre Antoine Lacassagne, Nice
| | - Erwan Gabiache
- Nuclear Medicine Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | | |
Collapse
|
24
|
Patil KS, Hajare AA, Manjappa AS, More HN, Disouza JI. Design, development, in silico and in vitro characterization of Docetaxel-loaded TPGS/ Pluronic F 108 mixed micelles for improved cancer treatment. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Horst EN, Bregenzer ME, Mehta P, Snyder CS, Repetto T, Yang-Hartwich Y, Mehta G. Personalized models of heterogeneous 3D epithelial tumor microenvironments: Ovarian cancer as a model. Acta Biomater 2021; 132:401-420. [PMID: 33940195 PMCID: PMC8969826 DOI: 10.1016/j.actbio.2021.04.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
Intractable human diseases such as cancers, are context dependent, unique to both the individual patient and to the specific tumor microenvironment. However, conventional cancer treatments are often nonspecific, targeting global similarities rather than unique drivers. This limits treatment efficacy across heterogeneous patient populations and even at different tumor locations within the same patient. Ultimately, this poor efficacy can lead to adverse clinical outcomes and the development of treatment-resistant relapse. To prevent this and improve outcomes, it is necessary to be selective when choosing a patient's optimal adjuvant treatment. In this review, we posit the use of personalized, tumor-specific models (TSM) as tools to achieve this remarkable feat. First, using ovarian cancer as a model disease, we outline the heterogeneity and complexity of both the cellular and extracellular components in the tumor microenvironment. Then we examine the advantages and disadvantages of contemporary cancer models and the rationale for personalized TSM. We discuss how to generate precision 3D models through careful and detailed analysis of patient biopsies. Finally, we provide clinically relevant applications of these versatile personalized cancer models to highlight their potential impact. These models are ideal for a myriad of fundamental cancer biology and translational studies. Importantly, these approaches can be extended to other carcinomas, facilitating the discovery of new therapeutics that more effectively target the unique aspects of each individual patient's TME. STATEMENT OF SIGNIFICANCE: In this article, we have presented the case for the application of biomaterials in developing personalized models of complex diseases such as cancers. TSM could bring about breakthroughs in the promise of precision medicine. The critical components of the diverse tumor microenvironments, that lead to treatment failures, include cellular- and extracellular matrix- heterogeneity, and biophysical signals to the cells. Therefore, we have described these dynamic components of the tumor microenvironments, and have highlighted how contemporary biomaterials can be utilized to create personalized in vitro models of cancers. We have also described the application of the TSM to predict the dynamic patterns of disease progression, and predict effective therapies that can produce durable responses, limit relapses, and treat any minimal residual disease.
Collapse
Affiliation(s)
- Eric N Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Michael E Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Catherine S Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Taylor Repetto
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, CT 06510, United States
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, United States; Precision Health, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
26
|
Potara M, Nagy-Simon T, Focsan M, Licarete E, Soritau O, Vulpoi A, Astilean S. Folate-targeted Pluronic-chitosan nanocapsules loaded with IR780 for near-infrared fluorescence imaging and photothermal-photodynamic therapy of ovarian cancer. Colloids Surf B Biointerfaces 2021; 203:111755. [PMID: 33862575 DOI: 10.1016/j.colsurfb.2021.111755] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/17/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022]
Abstract
Herein, we report the fabrication of a nanotherapeutic platform integrating near-infrared (NIR) imaging with combined therapeutic potential through photodynamic (PDT) and photothermal therapies (PTT) and recognition functionality against ovarian cancer. Owing to its NIR fluorescence, singlet oxygen generation and heating capacity, IR780 iodide is exploited to construct a multifunctional nanosystem for single-wavelength NIR laser imaging-assisted dual-modal phototherapy. We opted for loading IR780 into polymeric Pluronic-F127-chitosan nanoformulation in order to overcome its hydrophobicity and toxicity and to allow functionalization with folic acid. The obtained nanocapsules show temperature-dependent swelling and spectroscopic behavior with favorable size distribution for cellular uptake at physiological temperatures, improved fluorescence properties and good stability. The fabricated nanocapsules can efficiently generate singlet oxygen in solution and are able to produce considerable temperature increase (46 °C) upon NIR laser irradiation. Viability assays on NIH-OVCAR-3 cells confirm the successful biocompatibilization of IR780 by encapsulating in Pluronic and chitosan polymers. NIR fluorescence imaging assays reveal the ability of folic-acid functionalized nanocapsules to serve as intracellular contrast agents and demonstrate their active targeting capacity against folate receptor expressing ovarian cancer cells (NIH-OVCAR-3). Consequently, the targeted nanocapsules show improved NIR laser induced phototherapeutic performance against NIH-OVCAR-3 cells compared to free IR780. We anticipate that this class of nanocapsules holds great promise as theranostic agents for application in image-guided dual PDT-PTT and imaging assisted surgery of ovarian cancer.
Collapse
Affiliation(s)
- Monica Potara
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T Laurian 42, 400271, Cluj-Napoca, Romania
| | - Timea Nagy-Simon
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T Laurian 42, 400271, Cluj-Napoca, Romania
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T Laurian 42, 400271, Cluj-Napoca, Romania
| | - Emilia Licarete
- Molecular Biology Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T Laurian 42, 400271, Cluj-Napoca, Romania
| | - Olga Soritau
- Oncology Institute Prof. Dr. Ion Chiricuţă, 34-36 Republicii Street, 400015, Cluj-Napoca, Romania
| | - Adriana Vulpoi
- Nanostructured Materials and Bio-Nano-Interfaces Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T. Laurian 42, 400271, Cluj-Napoca, Romania
| | - Simion Astilean
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, T Laurian 42, 400271, Cluj-Napoca, Romania; Department of Biomolecular Physics, Faculty of Physics, Babes-Bolyai University, M Kogalniceanu 1, 400084, Cluj-Napoca, Romania.
| |
Collapse
|
27
|
Amobi-McCloud A, Muthuswamy R, Battaglia S, Yu H, Liu T, Wang J, Putluri V, Singh PK, Qian F, Huang RY, Putluri N, Tsuji T, Lugade AA, Liu S, Odunsi K. IDO1 Expression in Ovarian Cancer Induces PD-1 in T Cells via Aryl Hydrocarbon Receptor Activation. Front Immunol 2021; 12:678999. [PMID: 34025677 PMCID: PMC8136272 DOI: 10.3389/fimmu.2021.678999] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
The immunoregulatory enzyme, indoleamine 2,3-dioxygenase (IDO1) and the PD-1/PD-L1 axis are potent mechanisms that impede effective anti-tumor immunity in ovarian cancer. However, whether the IDO pathway regulates PD-1 expression in T cells is currently unknown. Here we show that tumoral IDO1 expression led to profound changes in tryptophan, nicotinate/nicotinamide, and purine metabolic pathways in the ovarian tumor microenvironment, and to an increased frequency of PD-1+CD8+ tumor infiltrating T cells. We determined that activation of the aryl hydrocarbon receptor (AHR) by kynurenine induced PD-1 expression, and this effect was significantly abrogated by the AHR antagonist CH223191. Mechanistically, kynurenine alters chromatin accessibility in regulatory regions of T cell inhibitory receptors, allowing AHR to bind to consensus XRE motifs in the promoter region of PD-1. These results enable the design of strategies to target the IDO1 and AHR pathways for enhancing anti-tumor immunity in ovarian cancer.
Collapse
Affiliation(s)
- Adaobi Amobi-McCloud
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Ravikumar Muthuswamy
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Han Yu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Tao Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jianmin Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Vasanta Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, United States
| | - Prashant K. Singh
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Feng Qian
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Ruea-Yea Huang
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nagireddy Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, United States
- Molecular and Cellular Biology, Advanced Technology Cores, Baylor College of Medicine, Houston, TX, United States
| | - Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Obstetrics and Gynecology-Gynecologic Oncology, University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, United States
| | - Amit A. Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Obstetrics and Gynecology-Gynecologic Oncology, University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, United States
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
28
|
Zhu H, Ai Y, Zhang J, Zhang J, Jin J, Xie C, Su H, Jin X. Preoperative Nomogram for Differentiation of Histological Subtypes in Ovarian Cancer Based on Computer Tomography Radiomics. Front Oncol 2021; 11:642892. [PMID: 33842352 PMCID: PMC8027335 DOI: 10.3389/fonc.2021.642892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/03/2021] [Indexed: 12/27/2022] Open
Abstract
Objectives Non-invasive method to predict the histological subtypes preoperatively is essential for the overall management of ovarian cancer (OC). The feasibility of radiomics in the differentiating of epithelial ovarian cancer (EOC) and non-epithelial ovarian cancer (NEOC) based on computed tomography (CT) images was investigated. Methods Radiomics features were extracted from preoperative CT for 101 patients with pathologically proven OC. Radiomics signature was built using the least absolute shrinkage and selection operator (LASSO) logistic regression. A nomogram was developed with the combination of radiomics features and clinical factors to differentiate EOC and NEOC. Results Eight radiomics features were selected to build a radiomics signature with an area under curve (AUC) of 0.781 (95% confidence interval (CI), 0.666 -0.897) in the discrimination between EOC and NEOC. The AUC of the combined model integrating clinical factors and radiomics features was 0.869 (95% CI, 0.783 -0.955). The nomogram demonstrated that the combined model provides a better net benefit to predict histological subtypes compared with radiomics signature and clinical factors alone when the threshold probability is within a range from 0.43 to 0.97. Conclusions Nomogram developed with CT radiomics signature and clinical factors is feasible to predict the histological subtypes preoperative for patients with OC.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Gynecology, Shanghai First Maternal and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Ai
- Department of Radiotherapy Center, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jindi Zhang
- Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ji Zhang
- Department of Radiotherapy Center, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Juebin Jin
- Department of Medical Engineering, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation and Medical Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Radiation and Medical Oncology, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huafang Su
- Department of Radiation and Medical Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiance Jin
- Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
29
|
Tong L, Cao W, Sheng J, Zhu E, Yu Y, Zhong T, Chen Y, Wang L. RDM1 plays an oncogenic role in human ovarian carcinoma cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:885-892. [PMID: 32501118 DOI: 10.1080/21691401.2020.1770267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Ovarian cancer is one of the deadliest gynecological cancer, with a low overall 5-year survival rate. RDM1, RAD52 motif-containing protein 1, is sensitive to cisplatin, a common chemotherapy drug and it has an important role inDNA damage repair pathway. Until now, the effect of RDM1 in ovarian cancer is undiscovered. Here, clinical data shows that the tumour tissues of ovarian carcinoma patients with higher mRNA and protein expression of RDM1. Knockdown of RDM1 in ovarian carcinoma cells reduces cell proliferation and promotes apoptosis, consistent with the role RDM1 in the overexpression experiments. The research of xenograft mouse model shows stable knockdown of RDM1 significantly inhibits ovarian cancer tumour growth. These in vitro and in vivo results conclude that RDM1 plays an oncogenic role in human ovarian carcinoma. Interestingly, p53/RAD51/RAD52 signalling pathway can be regulated by RDM1, and the negative regulation of p53 by RDM1 may be one of major mechanisms for RDM1 to accomplish its oncogenic functions in ovarian carcinoma. Therefore, RDM1 may be a new target for the treatment of ovarian carcinoma.
Collapse
Affiliation(s)
- Lu Tong
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, The China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenjiao Cao
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, The China Welfare Institute, Shanghai, China
| | - Jun Sheng
- Department of Orthopedics, the General Hospital of Western Theater Command, Chengdu, China
| | - Enhao Zhu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ying Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Tianying Zhong
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Lihua Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, The China Welfare Institute, Shanghai, China
| |
Collapse
|
30
|
In vivo selection of highly metastatic human ovarian cancer sublines reveals role for AMIGO2 in intra-peritoneal metastatic regulation. Cancer Lett 2021; 503:163-173. [PMID: 33524500 DOI: 10.1016/j.canlet.2021.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/06/2021] [Accepted: 01/23/2021] [Indexed: 01/06/2023]
Abstract
The majority of women with ovarian cancer are diagnosed with metastatic disease, therefore elucidating molecular events that contribute to successful metastatic dissemination may identify additional targets for therapeutic intervention and thereby positively impact survival. Using two human high grade serous ovarian cancer cell lines with inactive TP53 and multiple rounds of serial in vivo passaging, we generated sublines with significantly accelerated intra-peritoneal (IP) growth. Comparative analysis of the parental and IP sublines identified a common panel of differentially expressed genes. The most highly differentially expressed gene, upregulated by 60-65-fold in IP-selected sublines, was the type I transmembrane protein AMIGO2. As the role of AMIGO2 in ovarian cancer metastasis remains unexplored, CRISPR/Cas9 was used to reduce AMIGO2 expression, followed by in vitro and in vivo functional analyses. Knockdown of AMIGO2 modified the sphere-forming potential of ovarian cancer cells, reduced adhesion and invasion in vitro, and significantly attenuated IP metastasis. These data highlight AMIGO2 as a new target for a novel anti-metastatic therapeutic approach aimed at blocking cohesion, survival, and adhesion of metastatic tumorspheres.
Collapse
|
31
|
Yi X, Liu Y, Zhou B, Xiang W, Deng A, Fu Y, Zhao Y, Ouyang Q, Liu Y, Sun Z, Zhang K, Li X, Zeng F, Zhou H, Chen BT. Incorporating SULF1 polymorphisms in a pretreatment CT-based radiomic model for predicting platinum resistance in ovarian cancer treatment. Biomed Pharmacother 2021; 133:111013. [DOI: 10.1016/j.biopha.2020.111013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/03/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023] Open
|
32
|
Mysona DP, Tran L, Bai S, dos Santos B, Ghamande S, Chan J, She JX. Tumor-intrinsic and -extrinsic (immune) gene signatures robustly predict overall survival and treatment response in high grade serous ovarian cancer patients. Am J Cancer Res 2021; 11:181-199. [PMID: 33520368 PMCID: PMC7840710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/14/2020] [Indexed: 06/12/2023] Open
Abstract
In the present study, we developed a transcriptomic signature capable of predicting prognosis and response to primary therapy in high grade serous ovarian cancer (HGSOC). Proportional hazard analysis was performed on individual genes in the TCGA RNAseq data set containing 229 HGSOC patients. Ridge regression analysis was performed to select genes and develop multigenic models. Survival analysis identified 120 genes whose expression levels were associated with overall survival (OS) (HR = 1.49-2.46 or HR = 0.48-0.63). Ridge regression modeling selected 38 of the 120 genes for development of the final Ridge regression models. The consensus model based on plurality voting by 68 individual Ridge regression models classified 102 (45%) as low, 23 (10%) as moderate and 104 patients (45%) as high risk. The median OS was 31 months (HR = 7.63, 95% CI = 4.85-12.0, P < 1.0-10) and 77 months (HR = ref) in the high and low risk groups, respectively. The gene signature had two components: intrinsic (proliferation, metastasis, autophagy) and extrinsic (immune evasion). Moderate/high risk patients had more partial and non-responses to primary therapy than low risk patients (odds ratio = 4.54, P < 0.001). We concluded that the overall survival and response to primary therapy in ovarian cancer is best assessed using a combination of gene signatures. A combination of genes which combines both tumor intrinsic and extrinsic functions has the best prediction. Validation studies are warranted in the future.
Collapse
Affiliation(s)
- David P Mysona
- University of North CarolinaChapel Hill, NC 27517, USA
- Jinfiniti Precision Medicine, Inc.Augusta, GA 30907, USA
| | - Lynn Tran
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | | | - Sharad Ghamande
- Department of OBGYN, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| | - John Chan
- Palo Alto Medical Foundation Research InstitutePalo Alto, CA 94301, USA
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
- Department of OBGYN, Medical College of Georgia at Augusta UniversityAugusta, GA 30912, USA
| |
Collapse
|
33
|
Feng Y, Le F, Tian P, Zhong Y, Zhan F, Huang G, Hu H, Chen T, Tan B. GTW inhibits the Epithelial to Mesenchymal Transition of Epithelial Ovarian Cancer via ILK/AKT/GSK3β/Slug Signalling Pathway. J Cancer 2021; 12:1386-1397. [PMID: 33531984 PMCID: PMC7847657 DOI: 10.7150/jca.52418] [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: 08/25/2020] [Accepted: 12/09/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Epithelial ovarian cancer (EOC) accounts for the most lethal of all gynaecological cancers which is attributed to metastasis, invasiveness and drug resistance. A crucial link has been found between epithelial-mesenchymal transition (EMT) and cancer metastasis and chemo-resistance. Previous studies have confirmed that one of the main components of tripterygium glycosides (GTW)-triptolide (TPL) has anticancer effects. Methods: The purpose of this study is to determine whether GTW could inhibit EMT in A2780/DPP cells in vitro and in vivo, and explore the underlying mechanism. Results: In vitro results showed that GTW inhibited cell proliferation, invasion and migration, and intensified the sensitivity of A2780/DDP cells to cisplatin (DDP). GTW, especially GTW+DDP, significantly inhibited the expression of N-cadherin, integrin-linked kinase (ILK), phospho-protein kinase B/AKT (PKB/p-AKT), phospho-glycogen synthase kinase (p-GSK3β) and Slug, while it increased E-cadherin levels by inhibiting EMT via the ILK/AKT/GSK3β/Slug signalling pathway. Animal results indicated that GTW, especially GTW+DDP, significantly reduced tumour burden, prolonged the life span of mice, and down-regulated the levels of tumour markers CA125 and HE4 by regulating EMT through the ILK/AKT/GSK3β/Slug signalling pathway. Conclusion: Our results highlighted the significance of EMT in EOC metastasis, invasiveness and resistance to DDP and investigated the potential role of GTW as an adjuvant therapeutic agent in chemo-resistant EOC.
Collapse
Affiliation(s)
- Ying Feng
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Fuyin Le
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Puyuan Tian
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Yanying Zhong
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Fuliang Zhan
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Genhua Huang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Hui Hu
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Tingtao Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Buzhen Tan
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China
| |
Collapse
|
34
|
CA125 and Ovarian Cancer: A Comprehensive Review. Cancers (Basel) 2020; 12:cancers12123730. [PMID: 33322519 PMCID: PMC7763876 DOI: 10.3390/cancers12123730] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary CA125 has been the most promising biomarker for screening ovarian cancer; however, it still does not have an acceptable accuracy in population-based screening for ovarian cancer. In this review article, we have discussed the role of CA125 in diagnosis, evaluating response to treatment and prognosis of ovarian cancer and provided some suggestions in improving the clinical utility of this biomarker in the early diagnosis of aggressive ovarian cancers. These include using CA125 to screen individuals with symptoms who seek medical care rather than screening the general population, increasing the cutoff point for the CA125 level in the plasma and performing the test at point-of-care rather than laboratory testing. By these strategies, we would detect more aggressive ovarian cancer patients in stages that the tumour can be completely removed by surgery, which is the most important factor in redusing recurrence rate and improving the survival of the patients with ovarian cancer. Abstract Ovarian cancer is the second most lethal gynecological malignancy. The tumour biomarker CA125 has been used as the primary ovarian cancer marker for the past four decades. The focus on diagnosing ovarian cancer in stages I and II using CA125 as a diagnostic biomarker has not improved patients’ survival. Therefore, screening average-risk asymptomatic women with CA125 is not recommended by any professional society. The dualistic model of ovarian cancer carcinogenesis suggests that type II tumours are responsible for the majority of ovarian cancer mortality. However, type II tumours are rarely diagnosed in stages I and II. The recent shift of focus to the diagnosis of low volume type II ovarian cancer in its early stages of evolution provides a new and valuable target for screening. Type II ovarian cancers are usually diagnosed in advanced stages and have significantly higher CA125 levels than type I tumours. The detection of low volume type II carcinomas in stage IIIa/b is associated with a higher likelihood for optimal cytoreduction, the most robust prognostic indicator for ovarian cancer patients. The diagnosis of type II ovarian cancer in the early substages of stage III with CA125 may be possible using a higher cutoff point rather than the traditionally used 35 U/mL through the use of point-of-care CA125 assays in primary care facilities. Rapid point-of-care testing also has the potential for effective longitudinal screening and quick monitoring of ovarian cancer patients during and after treatment. This review covers the role of CA125 in the diagnosis and management of ovarian cancer and explores novel and more effective screening strategies with CA125.
Collapse
|
35
|
PD-L1 regulates tumorigenesis and autophagy of ovarian cancer by activating mTORC signaling. Biosci Rep 2020; 39:221398. [PMID: 31799599 PMCID: PMC6923342 DOI: 10.1042/bsr20191041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 01/09/2023] Open
Abstract
PD-L1 is a well-known immune co-stimulatory molecule that regulates tumour cell escape from immunity by suppressing the immune response. However, the clinical significance of PD-L1 in the progression of ovarian cancer is unclear. Our study demonstrated that PD-L1 is up-regulated in ovarian tumour tissue compared with its expression level in adjacent normal tissue. Furthermore, we confirmed that PD-L1 increases the proliferation of cancer cells by activating the AKT-mTORC signalling pathway, which is also enhanced by the expression of S6K, the substrate of mTORC. In addition, PD-L1 promotes the autophagy of ovarian cancer cells by up-regulating the expression of BECN1, a crucial molecule involved in the regulation of autophagy. In conclusion, PD-L1 may provide a target for the development of a novel strategy for the treatment of ovarian cancer.
Collapse
|
36
|
Endo H, Hama N, Baghdadi M, Ishikawa K, Otsuka R, Wada H, Asano H, Endo D, Konno Y, Kato T, Watari H, Tozawa A, Suzuki N, Yokose T, Takano A, Kato H, Miyagi Y, Daigo Y, Seino KI. Interleukin-34 expression in ovarian cancer: a possible correlation with disease progression. Int Immunol 2020; 32:175-186. [PMID: 31868884 DOI: 10.1093/intimm/dxz074] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 11/27/2019] [Indexed: 01/03/2023] Open
Abstract
Ovarian cancer is the second-most lethal gynecological malignancy and the seventh-commonest cause of cancer-related death in women around the world. Most of the ovarian cancer patients are diagnosed at advanced stages and suffer from recurrence after primary cytoreductive surgery and standard first-line chemotherapy. Thus, the successful management of ovarian cancer patients requires the identification of factors that contribute to progression and relapse. Interleukin-34 (IL-34) is a novel cytokine that acts as a tissue-specific ligand of colony-stimulating factor-1 receptor (CSF-1R). In cancer, IL-34 exerts pro-tumorigenic functions that promote tumor growth, metastasis, angiogenesis, immune suppression and therapeutic resistance. In this study, we evaluate the impact of IL-34 on progression and survival of ovarian cancer patients. First, IL-34 was found to be expressed in several human ovarian cancer cell lines and cancer tissues from patients. The expression of IL-34 was enhanced by cytotoxic chemotherapy in ovarian cancer cell lines and cancer tissues from chemotherapy-treated ovarian cancer patients. Importantly, high IL-34 expression correlated with worse progression-free survival (PFS) and overall survival in different cohorts. The assessment of PFS based on a combination between IL34 expression and other related genes such as CSF1R and CD163 helped further to reach more statistical significance compared with IL34 alone. Furthermore, in the murine ovarian cancer cell HM-1 in vivo model, it was suggested that IL-34-derived tumor cells was correlated with tumor progression and survival by modulating the immune environment. Collectively, these findings indicate a possible correlation between IL-34 expression and disease progression in ovarian cancer patients and the mouse model.
Collapse
Affiliation(s)
- Hiraku Endo
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki City, Kanagawa, Japan
| | - Naoki Hama
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Muhammad Baghdadi
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Kozo Ishikawa
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Otsuka
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Haruka Wada
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Asano
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daisuke Endo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yosuke Konno
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Kato
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akiko Tozawa
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki City, Kanagawa, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki City, Kanagawa, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Atsushi Takano
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Japan.,Center for Antibody and Vaccine, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hisamori Kato
- Department of Gynecology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center, Yokohama, Japan
| | - Yataro Daigo
- Department of Medical Oncology and Cancer Center, Shiga University of Medical Science, Otsu, Japan.,Center for Antibody and Vaccine, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
37
|
Shearsmith L, Kennedy F, Lindner OC, Velikova G. Delphi survey to inform patient-reported symptom monitoring after ovarian cancer treatment. J Patient Rep Outcomes 2020; 4:71. [PMID: 32857244 PMCID: PMC7453693 DOI: 10.1186/s41687-020-00237-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/18/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing numbers of ovarian cancer patients are living longer and requiring regular follow-up to detect disease recurrence. New models of follow-up care are needed to meet the growing number and needs of this patient group. The potential for patient-reported outcome measures (PROMs) to capture key symptoms and online technology to facilitate long-term follow-up has been suggested. OBJECTIVES Prior to a pilot study exploring the potential for electronic patient-reported symptom monitoring, the content of an online intervention was developed via Delphi methodology. DESIGN AND SETTING A Delphi process was conducted aiming to obtain consensus amongst the clinicians and patients from 4 hospitals on the key aspects to monitor during follow-up after ovarian cancer treatment, and how to monitor them in an online intervention. A two round Delphi was conducted. Consensus was defined as at least 70% agreement. RESULTS Out of 43 participants, 30 (18 patients, 12 healthcare professionals) completed round 1 and 19 (11 patients, 8 healthcare professionals) completed round 2. Consensus was reached on the key symptoms to monitor, and the importance of monitoring both duration and frequency of symptoms. Opportunity for review of psychological wellbeing and holistic needs were considered important by both groups. The frequency of online questionnaire completion, timeframe for patients to reflect on (e.g. during the past X weeks), and the choice of PROMs items to monitor symptoms did not reach the consensus threshold. CONCLUSION It is crucial that any intervention and the selection of PROMs is fully described to ensure transparency about the development and decisions taken. In this work, a set of key symptoms and areas to monitor were agreed, which has informed the design of an online intervention and a subsequent pilot study is now underway. The proposed model of remote follow-up using electronic PROMs could be adapted and explored in other cancer sites.
Collapse
Affiliation(s)
- Leanne Shearsmith
- Patient Reported Outcomes Group, Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, St James's Hospital, Level 6 Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Fiona Kennedy
- Patient Reported Outcomes Group, Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, St James's Hospital, Level 6 Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK.
| | - Oana C Lindner
- Patient Reported Outcomes Group, Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, St James's Hospital, Level 6 Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| | - Galina Velikova
- Patient Reported Outcomes Group, Section of Patient-Centred Outcomes Research, Leeds Institute of Medical Research (LIMR) at St James's, University of Leeds, St James's Hospital, Level 6 Bexley Wing, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
38
|
Yu KH, Hu V, Wang F, Matulonis UA, Mutter GL, Golden JA, Kohane IS. Deciphering serous ovarian carcinoma histopathology and platinum response by convolutional neural networks. BMC Med 2020; 18:236. [PMID: 32807164 PMCID: PMC7433108 DOI: 10.1186/s12916-020-01684-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/28/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Ovarian cancer causes 151,900 deaths per year worldwide. Treatment and prognosis are primarily determined by the histopathologic interpretation in combination with molecular diagnosis. However, the relationship between histopathology patterns and molecular alterations is not fully understood, and it is difficult to predict patients' chemotherapy response using the known clinical and histological variables. METHODS We analyzed the whole-slide histopathology images, RNA-Seq, and proteomics data from 587 primary serous ovarian adenocarcinoma patients and developed a systematic algorithm to integrate histopathology and functional omics findings and to predict patients' response to platinum-based chemotherapy. RESULTS Our convolutional neural networks identified the cancerous regions with areas under the receiver operating characteristic curve (AUCs) > 0.95 and classified tumor grade with AUCs > 0.80. Functional omics analysis revealed that expression levels of proteins participated in innate immune responses and catabolic pathways are associated with tumor grade. Quantitative histopathology analysis successfully stratified patients with different response to platinum-based chemotherapy (P = 0.003). CONCLUSIONS These results indicated the potential clinical utility of quantitative histopathology evaluation in tumor cell detection and chemotherapy response prediction. The developed algorithm is easily extensible to other tumor types and treatment modalities.
Collapse
Affiliation(s)
- Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA. .,Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Vincent Hu
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Feiran Wang
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George L Mutter
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Lysophosphatidic acid modulates ovarian cancer multicellular aggregate assembly and metastatic dissemination. Sci Rep 2020; 10:10877. [PMID: 32616784 PMCID: PMC7331713 DOI: 10.1038/s41598-020-67565-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) metastasis occurs by exfoliation of cells and multicellular aggregates (MCAs) from the tumor into the peritoneal cavity, adhesion to and retraction of peritoneal mesothelial cells and subsequent anchoring. Elevated levels of lysophosphatidic acid (LPA) have been linked to aberrant cell proliferation, oncogenesis, and metastasis. LPA disrupts junctional integrity and epithelial cohesion in vitro however, the fate of free-floating cells/MCAs and the response of host peritoneal tissues to LPA remain unclear. EOC MCAs displayed significant LPA-induced changes in surface ultrastructure with the loss of cell surface protrusions and poor aggregation, resulting in increased dissemination of small clusters compared to untreated control MCAs. LPA also diminished the adhesive capacity of EOC single cells and MCAs to murine peritoneal explants and impaired MCA survival and mesothelial clearance competence. Peritoneal tissues from healthy mice injected with LPA exhibited enhanced mesothelial surface microvilli. Ultrastructural alterations were associated with restricted peritoneal susceptibility to metastatic colonization by single cells as well as epithelial-type MCAs. The functional consequence is an LPA-induced dissemination of small mesenchymal-type clusters, promoting a miliary mode of peritoneal seeding that complicates surgical removal and is associated with worse prognosis.
Collapse
|
40
|
Zhao C, She X, Zhang Y, Liu C, Li P, Chen S, Sai B, Li Y, Feng J, Liu J, Sun Y, Xiao S, Li L, Wu M. LRRC4 Suppresses E-Cadherin-Dependent Collective Cell Invasion and Metastasis in Epithelial Ovarian Cancer. Front Oncol 2020; 10:144. [PMID: 32117780 PMCID: PMC7033568 DOI: 10.3389/fonc.2020.00144] [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] [Received: 08/23/2019] [Accepted: 01/27/2020] [Indexed: 11/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most malignant gynecological carcinoma and is of a high incidence of death due to detection at late stages when metastasis already occurs. However, the mechanism underlying metastasis of EOC remains unclear. Analysis of the open database and experiments with immunochemistry showed that LRRC4 is lowly expressed in high-grade serous ovarian cancer (HGSC) cells and during EOC metastasis. The 3D cell culture system and the orthotopic ovarian xenograft model infected with LRRC4-containing adeno-associated virus serotype 9 (AAV9) were used to confirm collective invasion and metastasis of cells in vitro and in vivo. Phos-tag SDS-PAGE was used to detect the phosphorylation of LRRC4 and PIK3R1. A number of experiments with methods such as co-immunoprecipitation and immunoblotting were performed to explore the mechanism for the actions of LRRC4 and PIK3R1 in EOC metastasis. An inverse correlation between LRRC4 and E-cadherin expression was detected in the regions of invasion in primary EOC tissues and metastatic ascites. LRRC4 binds to the cSH2 domain of PIK3R1 and inhibits the activity of PIK3R1, without disrupting the physical interactions between PIK3R1 and PIK3CA. LRRC4 inhibits EOC metastasis by targeting E-cadherin-dependent collective cell invasion and does so by inhibiting the PIK3R1-mediated AKT/GSK3β/β-catenin signaling pathway. LRRC4 functions as a tumor suppressor gene to inhibit EOC collective invasion and metastasis in vitro and in vivo and does so by directly binding to the cSH2 domain of PIK3R1 to exert its regulatory function. Our findings provide a potential novel approach for metastasis prognosis and a new strategy for the treatment of EOC.
Collapse
Affiliation(s)
- Chunhua Zhao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Xiaoling She
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| | - Shuai Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Buqing Sai
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| | - Yunchao Li
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| | - Jia Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Songshu Xiao
- Third Xiangya Hospital, Central South University, Changsha, China
| | - Liping Li
- The Affiliated Zhuzhou Hospital of XiangYa Medical School, Central South University, Changsha, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, China.,Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
| |
Collapse
|
41
|
Overcoming Resistance to Platinum-Based Drugs in Ovarian Cancer by Salinomycin and Its Derivatives-An In Vitro Study. Molecules 2020; 25:molecules25030537. [PMID: 31991882 PMCID: PMC7037477 DOI: 10.3390/molecules25030537] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/16/2022] Open
Abstract
Polyether ionophore salinomycin (SAL) and its semi-synthetic derivatives are recognized as very promising anticancer drug candidates due to their activity against various types of cancer cells, including multidrug-resistant populations. Ovarian cancer is the deadliest among gynecologic malignancies, which is connected with the development of chemoresistant forms of the disease in over 70% of patients after initial treatment regimen. Thus, we decided to examine the anticancer properties of SAL and selected SAL derivatives against a series of drug-sensitive (A2780, SK-OV-3) and derived drug-resistant (A2780 CDDP, SK-OV-3 CDDP) ovarian cancer cell lines. Although SAL analogs showed less promising IC50 values than SAL, they were identified as the antitumor agents that significantly overcome the resistance to platinum-based drugs in ovarian cancer, more potent than unmodified SAL and commonly used anticancer drugs—5-fluorouracil, gemcitabine, and cisplatin. Moreover, when compared with SAL used alone, our experiments proved for the first time increased selectivity of SAL-based dual therapy with 5-fluorouracil or gemcitabine, especially towards A2780 cell line. Looking closer at the results, SAL acted synergistically with 5-fluorouracil towards the drug-resistant A2780 cell line. Our results suggest that combinations of SAL with other antineoplastics may become a new therapeutic option for patients with ovarian cancer.
Collapse
|
42
|
Jo E, Jang HJ, Yang KE, Jang MS, Huh YH, Yoo HS, Park JS, Jang IS, Park SJ. Cordyceps militaris induces apoptosis in ovarian cancer cells through TNF-α/TNFR1-mediated inhibition of NF-κB phosphorylation. BMC Complement Med Ther 2020; 20:1. [PMID: 32020859 PMCID: PMC7076896 DOI: 10.1186/s12906-019-2780-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/29/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Cordyceps militaris (L.) Fr. (C. militaris) exhibits pharmacological activities, including antitumor properties, through the regulation of the nuclear factor kappa B (NF-κB) signaling. Tumor Necrosis Factor (TNF) and TNF-α modulates cell survival and apoptosis through NF- κB signaling. However, the mechanism underlying its mode of action on the NF-κB pathway is unclear. METHODS Here, we analyzed the effect of C. militaris extract (CME) on the proliferation of ovarian cancer cells by confirming viability, morphological changes, migration assay. Additionally, CME induced apoptosis was determined by apoptosis assay and apoptotic body formation under TEM. The mechanisms of CME were determined through microarray, immunoblotting and immunocytochemistry. RESULTS CME reduced the viability of cells in a dose-dependent manner and induced morphological changes. We confirmed the decrease in the migration activity of SKOV-3 cells after treatment with CME and the consequent induction of apoptosis. Immunoblotting results showed that the CME-mediated upregulation of tumor necrosis factor receptor 1 (TNFR1) expression induced apoptosis of SKOV-3 cells via the serial activation of caspases. Moreover, CME negatively modulated NF-κB activation via TNFR expression, suggestive of the activation of the extrinsic apoptotic pathway. The binding of TNF-α to TNFR results in the disassociation of IκB from NF-κB and the subsequent translocation of the active NF-κB to the nucleus. CME clearly suppressed NF-κB translocation induced by interleukin (IL-1β) from the cytosol into the nucleus. The decrease in the expression levels of B cell lymphoma (Bcl)-xL and Bcl-2 led to a marked increase in cell apoptosis. CONCLUSION These results suggest that C. militaris inhibited ovarian cancer cell proliferation, survival, and migration, possibly through the coordination between TNF-α/TNFR1 signaling and NF-κB activation. Taken together, our findings provide a new insight into a novel treatment strategy for ovarian cancer using C. militaris.
Collapse
Affiliation(s)
- Eunbi Jo
- Division of Analytical Science, Korea Basic Science Institute, Gwahangno 113, Yuseong-gu, Daejeon, 305-333, Republic of Korea
| | - Hyun-Jin Jang
- Division of Analytical Science, Korea Basic Science Institute, Gwahangno 113, Yuseong-gu, Daejeon, 305-333, Republic of Korea.,Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Eun Yang
- Division of Analytical Science, Korea Basic Science Institute, Gwahangno 113, Yuseong-gu, Daejeon, 305-333, Republic of Korea
| | - Min Su Jang
- Division of Biological Science and Technology, Yonsei University, Wonju, 220-100, Republic of Korea
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Hwa-Seung Yoo
- East-West Cancer Center, Daejeon University, Daejeon, 302-120, South Korea
| | - Jun Soo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, 220-100, Republic of Korea
| | - Ik-Soon Jang
- Division of Analytical Science, Korea Basic Science Institute, Gwahangno 113, Yuseong-gu, Daejeon, 305-333, Republic of Korea. .,Division of Analytical Science, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Soo Jung Park
- Department of Sasang Constitutional Medicine, College of Korean Medicine, Woosuk University, Wanju, Jeonbuk, 55338, Republic of Korea.
| |
Collapse
|
43
|
Alfaifi MY, Elbehairi SI, Shati AA, Fahmy UA, Alhakamy NA, Md S. Ellagic Acid Loaded TPGS Micelles for Enhanced Anticancer Activities in Ovarian Cancer. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2020.63.71] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Bae H, Jung DC, Lee JY, Nam EJ, Kang WJ, Oh YT. Patterns of initially overlooked recurrence of peritoneal lesions in patients with advanced ovarian cancer on postoperative multi-detector row CT. Acta Radiol 2019; 60:1713-1720. [PMID: 31081339 DOI: 10.1177/0284185119842832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Heejin Bae
- Department of Radiology, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dae Chul Jung
- Department of Radiology, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Ji Nam
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Taik Oh
- Department of Radiology, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Activin A promotes ovarian cancer cell migration by suppressing E-cadherin expression. Exp Cell Res 2019; 382:111471. [DOI: 10.1016/j.yexcr.2019.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/23/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
|
46
|
Gao B, Yang F, Chen W, Li R, Hu X, Liang Y, Li D. Multidrug resistance affects the prognosis of primary epithelial ovarian cancer. Oncol Lett 2019; 18:4262-4269. [PMID: 31579424 DOI: 10.3892/ol.2019.10745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 04/15/2019] [Indexed: 11/06/2022] Open
Abstract
Multidrug-resistant tumor cells can tolerate different structures, functions and antidrug action mechanisms, therefore, allowing these cells to respond to various structurally unrelated mechanisms of different chemotherapy drugs and to exhibit cross-resistance. The present study aimed to investigate the role of Multi-drug resistance gene (MDR1), Placental glutathione S-transferase-P1 (GSTP1), Lung resistance protein (LRP) and Ras association domain family member 1 (RASSF1A) in primary epithelial ovarian cancer (PEOC). The mRNA (protein) expression levels of MDR1, product P glycoprotein, LRP and GSTP1 were evaluated with reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis in all tissue samples, ovarian cancer cell line A2780 and A2780/DDP. Methylation-specific PCR (MSP) was used to detect RASSF1A gene methylation in all tissue samples. The resistance genes/proteins were either poorly or not expressed in A2780, however were highly expressed in A2780/DDP cell line. The expression of resistance genes/proteins decreased following different concentrations of zebularine-stimulated A2780/DDP. Hypermethylation and low expression of RASSF1A gene were detected in PEOC and A2780/DDP. Subsequent to being exposed to different concentrations of zebularine-stimulated A2780/DDP, the RASSF1A methylation level was decreased, while the unmethylation level was increased. The expression of RASSF1A gene/protein was gradually restored, and the gene/protein expression was enhanced with the increase in drug concentration. Multivariate logistic regression indicated that the expression level of gene LRP and GSTP1 was a risk factor for PEOC prognosis. Furthermore, the expression of LRP and GSTP1 in the negative-group survival curves was higher compared with the positive group. High expression of resistance genes may serve an important role in cancer primary resistance. Low expression caused by hyper-methylation of RASSF1A gene may serve an important role in cancer-acquired resistance in PEOC. The present study suggested that resistant gene expression may be a potential prognostic biomarker.
Collapse
Affiliation(s)
- Bo Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi 710061, P.R. China.,Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Fengmei Yang
- Department of Obstetrics and Gynecology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wei Chen
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Rui Li
- Department of Medical Office, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiuxue Hu
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yong Liang
- Department of Anesthesiology, Ren-ming Hospital of Yun-xi, Shiyan, Hubei 442000, P.R. China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi 710061, P.R. China
| |
Collapse
|
47
|
Feng X, Bai X, Ni J, Wasinger VC, Beretov J, Zhu Y, Graham P, Li Y. CHTOP in Chemoresistant Epithelial Ovarian Cancer: A Novel and Potential Therapeutic Target. Front Oncol 2019; 9:557. [PMID: 31380263 PMCID: PMC6660285 DOI: 10.3389/fonc.2019.00557] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023] Open
Abstract
Objective: Chemoresistance is a major challenge in epithelial ovarian cancer (EOC) treatment. Chromatin target of protein arginine methyltransferase (CHTOP) was identified as a potential biomarker in chemoresistant EOC cell lines using label-free LC-MS/MS quantitative proteomics. Thus, the aim of this study is to investigate the role of CHTOP in chemoresistant EOC and the underlying mechanism. Methods: The expression of CHTOP in human ovarian cancer cells and tissues was detected using immunofluorescence (IF), western blot (WB), and immunohistochemistry (IHC), respectively. Flow cytometry and TUNEL assay were employed to detect the effect of CHTOP knockdown (KD) in chemoresistant EOC cell apoptosis, while colony and sphere formation assays were used to evaluate its effect on cell stemness. The association of CHTOP with cell metastasis was determined using Matrigel invasion and wound-healing assays. Results: The higher level expression of CHTOP protein was found in chemoresistant EOC cells as compared to their sensitive parental cells or normal epithelial ovarian cells. Results from IHC and bioinformatic analysis showed CHTOP was highly expressed in human ovarian cancer tissues and associated with a poor progression-free survival in patients. In addition, CHTOP KD significantly enhanced cisplatin-induced apoptosis, reduced the stemness of chemoresistant EOC cells, and decreased their metastatic potential. Conclusion: Our findings suggest that CHTOP is associated with apoptosis, stemness, and metastasis in chemoresistant EOC cells and might be a promising target to overcome chemoresistance in EOC treatment.
Collapse
Affiliation(s)
- Xiaojie Feng
- Department of Gynaecological Oncology, Henan Cancer Hospital, Zhengzhou, China.,Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Xupeng Bai
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Jie Ni
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Valerie C Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Sydney, NSW, Australia.,School of Medical Science, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Julia Beretov
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia.,Anatomical Pathology, NSW Health Pathology, St. George Hospital, Kogarah, NSW, Australia
| | - Ying Zhu
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Peter Graham
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Yong Li
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia.,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
48
|
Chen D, Cao L, Wang X. MPZL1 promotes tumor cell proliferation and migration via activation of Src kinase in ovarian cancer. Oncol Rep 2019; 42:679-687. [PMID: 31233194 PMCID: PMC6610034 DOI: 10.3892/or.2019.7199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor metastasis is the leading cause of mortality in patients with advanced ovarian cancer. Myelin protein zero like 1 (MPZL1) is a transmembrane glycoprotein that promotes migration of hepatocellular carcinoma cells and is involved in extracellular matrix-induced signal transduction. However, the functional role of MPZL1 in ovarian cancer has not been well elucidated. The present study conducted western blotting, phase-contrast imaging and immunohistochemistry to reveal the functions of MPZL1 in ovarian cancer. The present study demonstrated that the expression levels of MPZL1 were associated with malignant features of ovarian cancer. Furthermore, overexpression of MPZL1 significantly promoted cell proliferation, migration and invasion of ovarian cancer cells. Conversely, MPZL1 depletion by short hairpin RNA inhibited migration and invasion of ovarian cancer cells. In addition, this study demonstrated that phosphorylation of Src kinase was increased upon MPZL1 overexpression. Additionally, phosphorylation and activation of pro-metastatic proteins p130 and cortactin were induced by phosphorylated Src kinase. Collectively, these findings indicated that MPZL1 may be a novel pro-metastatic gene, which promotes tumor cell proliferation and migration through Src-mediated phosphorylation of p130 and cortactin in ovarian cancer.
Collapse
Affiliation(s)
- Danni Chen
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Lei Cao
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Xiaojie Wang
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
49
|
Stasenko M, Cybulska P, Feit N, Makker V, Konner J, O'Cearbhaill RE, Alektiar KM, Beal K, Gardner GJ, Long Roche KC, Sonoda Y, Chi DS, Zivanovic O, Leitao MM, Cadoo KA, Tew WP. Brain metastasis in epithelial ovarian cancer by BRCA1/2 mutation status. Gynecol Oncol 2019; 154:144-149. [PMID: 31113680 DOI: 10.1016/j.ygyno.2019.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To evaluate clinical outcomes of patients with BRCA-associated ovarian cancer who developed brain metastases (BM). METHODS Patients with epithelial ovarian, fallopian tube, and primary peritoneal cancer (EOC) and BM, treated at a single institution from 1/1/2008-7/1/2018, were identified from two institutional databases. Charts and medical records were retrospectively reviewed for clinical characteristics and germline BRCA mutation status. Appropriate statistics were used. RESULTS Of 3649 patients with EOC, 91 had BM (2.5%). Germline mutation status was available for 63 (69%) cases; 21 (35%) of these harbored a BRCA1/2 mutation (15 BRCA1, 6 BRCA2). Clinical characteristics were similar between groups. BM were diagnosed at a median of 31 months (95% CI, 22.6-39.4) in BRCA-mutated (mBRCA) and 32 months (95% CI, 23.7-40.3) in wild-type BRCA (wtBRCA) (p = 0.78) patients. Brain metastases were the only evidence of disease at time of BM diagnoses in 48% (n = 10) mBRCA and 19% (n = 8) wtBRCA (p = 0.02) patients. There was no difference in treatment of BM by mutation status (p = 0.84). Survival from time of BM diagnosis was 29 months (95%CI, 15.5-42.5) in mBRCA and 9 months (95% CI, 5.5-12.5) in wtBRCA patients, with an adjusted hazard ratio (HR) of 0.53, p = 0.09; 95% CI, 0.25-1.11. HR was adjusted for presence of systemic disease at time of BM diagnosis. CONCLUSION This is the largest study to date comparing outcomes in patients with EOC and BM by mutation status. mBRCA patients were more likely to have isolated BM, which may be a factor in their long survival. This supports the pursuit of aggressive treatment for mBRCA EOC patients with BM. Additional studies examining the correlation of BRCA mutational status with BM are warranted.
Collapse
Affiliation(s)
- Marina Stasenko
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paulina Cybulska
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Noah Feit
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Vicky Makker
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason Konner
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Roisin E O'Cearbhaill
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kaled M Alektiar
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kathryn Beal
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ginger J Gardner
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Kara C Long Roche
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Yukio Sonoda
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Dennis S Chi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Oliver Zivanovic
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Mario M Leitao
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Karen A Cadoo
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - William P Tew
- Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
50
|
Chromatin target of protein arginine methyltransferase regulates invasion, chemoresistance, and stemness in epithelial ovarian cancer. Biosci Rep 2019; 39:BSR20190016. [PMID: 30910850 PMCID: PMC6465198 DOI: 10.1042/bsr20190016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/08/2019] [Accepted: 03/23/2019] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is one of the most common gynecological cancers with a high mortality rate in females. Chromatin target of protein arginine methyltransferase (CHTOP) is an important intracellular protein that regulates the transcriptional activation of several oncogenic genes in glioblastomagenesis and controls mature mRNA export as a component of TRanscription-Export complex. However, the role of CHTOP in ovarian cancer is unclear. In the present study, we investigated the correlation between tumor-derived CHTOP expression and prognosis and explored its role in the malignant behaviors of epithelial ovarian cancer cells. We found that higher expression of CHTOP was associated with a lower disease-free survival (DFS) rate in ovarian cancer patients. Also, CHTOP was highly expressed in human ovarian cancer tissues compared with normal and adjacent tissues. Moreover, compared with IGROV-1 cell line, higher expression of CHTOP was also confirmed in the malignant ovarian cancer cell lines (OV-90 and SK-OV-3). Further results from wound-healing and Matrigel assay showed that CHTOP knockdown significantly reduced the migration and invasion ability of OV-90 and SK-OV-3 cells, while colony formation assay and apoptosis detection showed that CHTOP knockdown markedly sensitized OV-90 and SK-OV-3 cells to cisplatin treatment by inducing apoptosis. Additionally, CHTOP silence also remarkably weakened the stemness of OV-90 and SK-OV-3 through inhibiting the protein expressions of several transcriptional or surface markers of cancer stem cells. These findings first suggest that CHTOP, as a highly expressed protein in ovarian cancer, is closely associated with the malignant phenotypes of epithelial ovarian cancer cells, including metastasis, chemoresistance, and stemness, which highlights a promising role of CHTOP in ovarian cancer targeted therapy.
Collapse
|