1
|
Segal BH, Giridharan T, Suzuki S, Khan ANH, Zsiros E, Emmons TR, Yaffe MB, Gankema AAF, Hoogeboom M, Goetschalckx I, Matlung HL, Kuijpers TW. Neutrophil interactions with T cells, platelets, endothelial cells, and of course tumor cells. Immunol Rev 2023; 314:13-35. [PMID: 36527200 PMCID: PMC10174640 DOI: 10.1111/imr.13178] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Thejaswini Giridharan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anm Nazmul H Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tiffany R Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela A F Gankema
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogeboom
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Ines Goetschalckx
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children's Hospital Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2
|
YILDIZ GÖ, SERTCAKACİLAR G, AKYOL D, KARAKAŞ S, HERGÜNSEL GO. Malign asitli over kanserinde sitoredüktif cerrahide perioperatif hemodinamik optimizasyon. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1097476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: In this study, we aimed to evaluate the effects of norepinephrine and albumin use in patients with epithelial ovarian cancer with malignant ascite in order to maintain plasma oncotic pressure and intravascular volume, to provide perioperative hemodynamic stabilization and tissue perfusion. In addition, it was aimed to compare in terms of postoperative intensive care admission, hospital stay and complications.
Materials and Methods: A total of 66 patients, 38 with ascites and 28 without ascites, who underwent cytoreductive surgery for ovarian cancer were included in this study. PVI and invasive arterial monitoring of the patients were performed after hemodynamic stabilization (after the start of surgery) (T0). T0, 1st hour (T1) and 2nd hour (T2) and postoperative (Tpostop.) Ascites patients were composed of 3 subgroups which the ones received norepinephrine (NE) infusion, norepinephrine + albumin (NEA) infusion or only fluid therapy (FT). From the perioperative hemodynamic and laboratory data of the patients, tissue perfusion was evaluated with lactate, and hemodynamic status was evaluated with pleth variability index (PVI), perfusion index (PI) and mean arterial pressure (MAP).
Results: Demographic and clinical findings did not differ significantly between patients with and without ascites. Lactate level in NEA / NE group in Tpostop, PVI level in T1h, T2h and Tpostop time frames were determined higher than the FT group. PI was found to be significantly lower in the T2 time frame. The postoperative ICU admission rate was higher in the NEA and NE groups. The duration of ICU stay in group NEA was shorter than in group NE.
Conclusion: We recommend the use of low-dose NE with albumin to provide perioperative hemodynamic optimization, tissue perfusion and plasma oncotic pressure in surgery of ovarian cancer with malignant ascites. Despite high fluid replacement in these patients, the use of norepinephrine and albumin together may have an important role in preventing / reducing major complications in the perioperative period.
Collapse
Affiliation(s)
- Güneş Özlem YILDIZ
- Department of Anesthesiology and Intensive Care, University of Health Sciences Istanbul, Bakirkoy Dr. Sadi Konuk Training and Research Hospital
| | - Gokhan SERTCAKACİLAR
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL BAKIRKÖY DR. SADİ KONUK TRAINING RESEARCH CENTER
| | - Duygu AKYOL
- SAĞLIK BİLİMLERİ ÜNİVERSİTESİ, İSTANBUL BAŞAKŞEHİR ÇAM VE SAKURA ŞEHİR SAĞLIK UYGULAMA VE ARAŞTIRMA MERKEZİ
| | - Sema KARAKAŞ
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL BAKIRKÖY DR. SADİ KONUK TRAINING RESEARCH CENTER
| | - Gülsüm Oya HERGÜNSEL
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL BAKIRKÖY DR. SADİ KONUK TRAINING RESEARCH CENTER
| |
Collapse
|
3
|
Bortot B, Apollonio M, Rampazzo E, Valle F, Brucale M, Ridolfi A, Ura B, Addobbati R, Di Lorenzo G, Romano F, Buonomo F, Ripepi C, Ricci G, Biffi S. Small extracellular vesicles from malignant ascites of patients with advanced ovarian cancer provide insights into the dynamics of the extracellular matrix. Mol Oncol 2021; 15:3596-3614. [PMID: 34614287 PMCID: PMC8637559 DOI: 10.1002/1878-0261.13110] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
The exact role of malignant ascites in the development of intraperitoneal metastases remains unclear, and the mechanisms by which extracellular vesicles (EVs) promote tumor progression in the pre-metastatic niche have not been fully discovered. In this study, we characterized ascites from high-grade epithelial ovarian cancer patients. Small-EVs (30-150 nm) were isolated from two sources-the bulk ascites and the ascitic fluid-derived tumor cell cultures-and assessed with a combination of imaging, proteomic profiling, and protein expression analyses. In addition, Gene Ontology and pathway analysis were performed using different databases and bioinformatic tools. The results proved that the small-EVs derived from the two sources exhibited significantly different stiffness and size distributions. The bulk ascitic fluid-derived small-EVs were predominantly involved in the complement and coagulation cascade. Small-EVs derived from ascites cell cultures contained a robust proteomic profile of extracellular matrix remodeling regulators, and we observed an increase in transforming growth factor-β-I (TGFβI), plasminogen activator inhibitor 1 (PAI-1), and fibronectin expression after neoadjuvant chemotherapy. When measured in the two sources, we demonstrated that fibronectin exhibited opposite expression patterns in small-EVs in response to chemotherapy. These findings highlight the importance of an ascites cell isolation workflow in investigating the treatment-induced cancer adaption processes.
Collapse
Affiliation(s)
- Barbara Bortot
- Department of Medical Genetics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Maura Apollonio
- Pediatric Department, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Enrico Rampazzo
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Italy
| | - Francesco Valle
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati (CNRISMN), Bologna, Italy
| | - Marco Brucale
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati (CNRISMN), Bologna, Italy
| | - Andrea Ridolfi
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Department of Chemistry, University of Firenze, Italy
| | - Blendi Ura
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Riccardo Addobbati
- Department of Clinical Toxicology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giovanni Di Lorenzo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Federico Romano
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Francesca Buonomo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Chiara Ripepi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.,Clinical Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Stefania Biffi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
4
|
Záveský L, Jandáková E, Weinberger V, Hanzíková V, Slanař O, Kohoutová M. Ascites in ovarian cancer: MicroRNA deregulations and their potential roles in ovarian carcinogenesis. Cancer Biomark 2021; 33:1-16. [PMID: 34511487 DOI: 10.3233/cbm-210219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ovarian cancer comprises the most lethal gynecologic malignancy and is accompanied by the high potential for the incidence of metastasis, recurrence and chemotherapy resistance, often associated with a formation of ascitic fluid. The differentially expressed ascites-derived microRNAs may be linked to ovarian carcinogenesis. The article focuses on a number of miRNAs that share a common expression pattern as determined by independent studies using ascites samples and with regard to their functions and outcomes in experimental and clinical investigations.Let-7b and miR-143 have featured as tumor suppressors in ovarian cancer, which is in line with data on other types of cancer. Although two miRNAs, i.e. miR-26a-5p and miR-145-5p, act principally as tumor suppressor miRNAs, they occasionally exhibit oncogenic roles. The performance of miR-95-3p, upregulated in ascites, is open to debate given the current lack of supportive data on ovarian cancer; however, data on other cancers indicates its probable oncogenic role. Different findings have been reported for miR-182-5p and miR-200c-3p; in addition to their presumed oncogenic roles, contrasting findings have indicated their ambivalent functions. Further research is required for the identification and evaluation of the potential of specific miRNAs in the diagnosis, prediction, treatment and outcomes of ovarian cancer patients.
Collapse
Affiliation(s)
- Luděk Záveský
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic.,Institute of Pharmacology, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| | - Eva Jandáková
- Department of Pathology, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Vít Weinberger
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Veronika Hanzíková
- Faculty Transfusion Center, General University Hospital, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| | - Milada Kohoutová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| |
Collapse
|
5
|
Croft PKD, Sharma S, Godbole N, Rice GE, Salomon C. Ovarian-Cancer-Associated Extracellular Vesicles: Microenvironmental Regulation and Potential Clinical Applications. Cells 2021; 10:cells10092272. [PMID: 34571921 PMCID: PMC8471580 DOI: 10.3390/cells10092272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is one of the most diagnosed gynecological cancers in women. Due to the lack of effective early stage screening, women are more often diagnosed at an advanced stage; therefore, it is associated with poor patient outcomes. There are a lack of tools to identify patients at the highest risk of developing this cancer. Moreover, early detection strategies, therapeutic approaches, and real-time monitoring of responses to treatment to improve survival and quality of life are also inadequate. Tumor development and progression are dependent upon cell-to-cell communication, allowing cancer cells to re-program cells not only within the surrounding tumor microenvironment, but also at distant sites. Recent studies established that extracellular vesicles (EVs) mediate bi-directional communication between normal and cancerous cells. EVs are highly stable membrane vesicles that are released from a wide range of cells, including healthy and cancer cells. They contain tissue-specific signaling molecules (e.g., proteins and miRNA) and, once released, regulate target cell phenotypes, inducing a pro-tumorigenic and immunosuppressive phenotype to contribute to tumor growth and metastasis as well as proximal and distal cell function. Thus, EVs are a “fingerprint” of their cell of origin and reflect the metabolic status. Additionally, via the capacity to evade the immune system and remain stable over long periods in circulation, EVs can be potent therapeutic agents. This review examines the potential role of EVs in the different aspects of the tumor microenvironment in OC, as well as their application in diagnosis, delivery of therapeutic agents, and disease monitoring.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia
| | - Shayna Sharma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Nihar Godbole
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Gregory E. Rice
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Correspondence: ; Tel.: +61-7-3346-5500; Fax: +61-7-3346-5509
| |
Collapse
|
6
|
Morgan RD, McNeish IA, Cook AD, James EC, Lord R, Dark G, Glasspool RM, Krell J, Parkinson C, Poole CJ, Hall M, Gallardo-Rincón D, Lockley M, Essapen S, Summers J, Anand A, Zachariah A, Williams S, Jones R, Scatchard K, Walther A, Kim JW, Sundar S, Jayson GC, Ledermann JA, Clamp AR. Objective responses to first-line neoadjuvant carboplatin-paclitaxel regimens for ovarian, fallopian tube, or primary peritoneal carcinoma (ICON8): post-hoc exploratory analysis of a randomised, phase 3 trial. Lancet Oncol 2021; 22:277-288. [PMID: 33357510 PMCID: PMC7616995 DOI: 10.1016/s1470-2045(20)30591-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Platinum-based neoadjuvant chemotherapy followed by delayed primary surgery (DPS) is an established strategy for women with newly diagnosed, advanced-stage epithelial ovarian cancer. Although this therapeutic approach has been validated in randomised, phase 3 trials, evaluation of response to neoadjuvant chemotherapy using Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST), and cancer antigen 125 (CA125) has not been reported. We describe RECIST and Gynecologic Cancer InterGroup (GCIG) CA125 responses in patients receiving platinum-based neoadjuvant chemotherapy followed by DPS in the ICON8 trial. METHODS ICON8 was an international, multicentre, randomised, phase 3 trial done across 117 hospitals in the UK, Australia, New Zealand, Mexico, South Korea, and Ireland. The trial included women aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0-2, life expectancy of more than 12 weeks, and newly diagnosed International Federation of Gynecology and Obstetrics (FIGO; 1988) stage IC-IIA high-grade serous, clear cell, or any poorly differentiated or grade 3 histological subtype, or any FIGO (1988) stage IIB-IV epithelial cancer of the ovary, fallopian tube, or primary peritoneum. Patients were randomly assigned (1:1:1) to receive intravenous carboplatin (area under the curve [AUC]5 or AUC6) and intravenous paclitaxel (175 mg/m2 by body surface area) on day 1 of every 21-day cycle (control group; group 1); intravenous carboplatin (AUC5 or AUC6) on day 1 and intravenous dose-fractionated paclitaxel (80 mg/m2 by body surface area) on days 1, 8, and 15 of every 21-day cycle (group 2); or intravenous dose-fractionated carboplatin (AUC2) and intravenous dose-fractionated paclitaxel (80 mg/m2 by body surface area) on days 1, 8, and 15 of every 21-day cycle (group 3). The maximum number of cycles of chemotherapy permitted was six. Randomisation was done with a minimisation method, and patients were stratified according to GCIG group, disease stage, and timing and outcome of cytoreductive surgery. Patients and clinicians were not masked to group allocation. The scheduling of surgery and use of neoadjuvant chemotherapy were determined by local multidisciplinary case review. In this post-hoc exploratory analysis of ICON8, progression-free survival was analysed using the landmark method and defined as the time interval between the date of pre-surgical planning radiological tumour assessment to the date of investigator-assessed clinical or radiological progression or death, whichever occurred first. This definition is different from the intention-to-treat primary progression-free survival analysis of ICON8, which defined progression-free survival as the time from randomisation to the date of first clinical or radiological progression or death, whichever occurred first. We also compared the extent of surgical cytoreduction with RECIST and GCIG CA125 responses. This post-hoc exploratory analysis includes only women recruited to ICON8 who were planned for neoadjuvant chemotherapy followed by DPS and had RECIST and/or GCIG CA125-evaluable disease. ICON8 is closed for enrolment and follow-up, and registered with ClinicalTrials.gov, NCT01654146. FINDINGS Between June 6, 2011, and Nov 28, 2014, 1566 women were enrolled in ICON8, of whom 779 (50%) were planned for neoadjuvant chemotherapy followed by DPS. Median follow-up was 29·5 months (IQR 15·6-54·3) for the neoadjuvant chemotherapy followed by DPS population. Of 564 women who had RECIST-evaluable disease at trial entry, 348 (62%) had a complete or partial response. Of 727 women who were evaluable by GCIG CA125 criteria at the time of diagnosis, 610 (84%) had a CA125 response. Median progression-free survival was 14·4 months (95% CI 9·2-28·0; 297 events) for patients with a RECIST complete or partial response and 13·3 months (8·1-20·1; 171 events) for those with RECIST stable disease. Median progression-free survival for women with a GCIG CA125 response was 13·8 months (95% CI 8·8-23·4; 544 events) and 9·7 months (5·8-14·5; 111 events) for those without a GCIG CA125 response. Complete cytoreduction (R0) was achieved in 187 (56%) of 335 women with a RECIST complete or partial response and 73 (42%) of 172 women with RECIST stable disease. Complete cytoreduction was achieved in 290 (50%) of 576 women with a GCIG CA125 response and 30 (30%) of 101 women without a GCIG CA125 response. INTERPRETATION The RECIST-defined radiological response rate was lower than that frequently quoted to patients in the clinic. RECIST and GCIG CA125 responses to neoadjuvant chemotherapy for epithelial ovarian cancer should not be used as individual predictive markers to stratify patients who are likely to benefit from DPS, but instead used in conjunction with the patient's clinical capacity to undergo cytoreductive surgery. A patient should not be denied surgery based solely on the lack of a RECIST or GCIG CA125 response. FUNDING Cancer Research UK, UK Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, and Cancer Australia.
Collapse
Affiliation(s)
- Robert D Morgan
- The Christie NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Iain A McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Adrian D Cook
- Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Elizabeth C James
- Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Rosemary Lord
- The Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, UK
| | - Graham Dark
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Jonathan Krell
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Christopher J Poole
- Arden Cancer Research Centre, University Hospital Coventry and Warwickshire NHS Trust, Coventry, UK
| | | | | | | | | | - Jeff Summers
- Maidstone and Tunbridge Wells NHS Trust, Kent, UK
| | - Anjana Anand
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Abel Zachariah
- Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, UK
| | - Sarah Williams
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rachel Jones
- South West Wales Cancer Centre, Singleton Hospital, Swansea, UK
| | | | - Axel Walther
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Jae-Weon Kim
- Seoul National University College of Medicine, Seoul, South Korea
| | - Sudha Sundar
- Pan Birmingham Gynaecological Cancer Centre and University of Birmingham, Birmingham, UK
| | - Gordon C Jayson
- The Christie NHS Foundation Trust and University of Manchester, Manchester, UK
| | | | - Andrew R Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, UK.
| |
Collapse
|
7
|
Biologically Active Tissue Factor-Bearing Larger Ectosome-Like Extracellular Vesicles in Malignant Effusions from Ovarian Cancer Patients: Correlation with Incidence of Thrombosis. Int J Mol Sci 2021; 22:ijms22020790. [PMID: 33466775 PMCID: PMC7829758 DOI: 10.3390/ijms22020790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The development of malignant effusions such as ascites reflects a massive progression of a malignant disease. In patients with ovarian carcinoma, a high amount of ascites (>500 mL) is an independent negative prognostic marker. The composition and constituents of ascites reflect the inflammatory environment of the underlying tumor. Increased cellular resistance of ascites-derived tumor cells and the development of venous thromboembolic events (VTE) are major risks for these patients, especially in patients with advanced ovarian carcinoma. In this study, we discuss the release of tissue factor-bearing extracellular vesicles (TF+ EVs) from tumor cells into the environment (ascites fluid) and their systemic spreading as a possible causal explanation of the pathologic coagulation status in these patients. We obtained ascites from patients with advanced ovarian carcinoma, collected during surgery or therapeutic paracentesis (n = 20). Larger ectosome-like EVs were isolated using sequential centrifugation, quantified by high-resolution flow cytometry and analyzed using nanoparticle tracking analysis. Furthermore, the pro-coagulant properties (TF activity) of EVs were determined. Compared to published TF activities of EVs from healthy persons, TF activities of EVs derived from ascites of patients with ovarian cancer were very high, with a median of 80 pg/mL. The rate of VTE, as reported in the patient files, was high as well (35%, 7 out of 20). Furthermore, all but one patient with VTE had EV concentrations above the median within their ascetic fluid (p < 0.02). Since VTE continues to be a frequent cause of death in cancer patients, prophylactic antithrombotic treatment might be worth considering in these patients. However, given the risk of bleeding, more clinical data are warranted. Although the study is too small to enable reaching a conclusion on direct clinical implementation, it can well serve as a proof of principle and a rationale to initiate a prospective clinical study with different patient subgroups. We also show ex vivo that these larger ectosome-like EVs induce intracellular ERK phosphorylation and tumor cell migration, which is not directly related to their pro-coagulative potency, but might help to understand why cancer patients with thromboembolic events have a poorer prognosis.
Collapse
|
8
|
Notch3 signaling promotes tumor cell adhesion and progression in a murine epithelial ovarian cancer model. PLoS One 2020; 15:e0233962. [PMID: 32525899 PMCID: PMC7289394 DOI: 10.1371/journal.pone.0233962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 05/15/2020] [Indexed: 11/26/2022] Open
Abstract
High grade serous ovarian cancer (HGSC) is the most common and deadly type of ovarian cancer, largely due to difficulties in early diagnosis and rapid metastasis throughout the peritoneal cavity. Previous studies have shown that expression of Notch3 correlates with worse prognosis and increased tumorigenic cell behaviors in HGSC. We investigated the mechanistic role of Notch3 in a model of metastatic ovarian cancer using the murine ovarian surface epithelial cell line, ID8 IP2. Notch3 was activated in ID8 IP2 cells via expression of the Notch3 intracellular domain (Notch3IC). Notch3IC ID8 IP2 cells injected intraperitoneally caused accelerated ascites and reduced survival compared to control ID8 IP2, particularly in early stages of disease. We interrogated downstream targets of Notch3IC in ID8 IP2 cells by RNA sequencing and found significant induction of genes that encode adhesion and extracellular matrix proteins. Notch3IC ID8 IP2 showed increased expression of ITGA1 mRNA and cell-surface protein. Notch3IC-mediated increase of ITGA1 was also seen in two human ovarian cancer cells. Notch3IC ID8 IP2 cells showed increased adhesion to collagens I and IV in vitro. We propose that Notch3 activation in ovarian cancer cells causes increased adherence to collagen-rich peritoneal surfaces. Thus, the correlation between increased Notch3 signaling and poor prognosis may be influenced by increased metastasis of HGSC via increased adherence of disseminating cells to new metastatic sites in the peritoneum.
Collapse
|
9
|
The Capacity of High-Grade Serous Ovarian Cancer Cells to Form Multicellular Structures Spontaneously along Disease Progression Correlates with Their Orthotopic Tumorigenicity in Immunosuppressed Mice. Cancers (Basel) 2020; 12:cancers12030699. [PMID: 32188032 PMCID: PMC7140084 DOI: 10.3390/cancers12030699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 12/16/2022] Open
Abstract
Many studies have examined the biology, genetics, and chemotherapeutic response of ovarian cancer's solid component; its liquid facet, however, remains critically underinvestigated. Floating within peritoneal effusions known as ascites, ovarian cancer cells form multicellular structures, creating a cancer niche in suspension. This study explores the pathobiology of spontaneously formed, multicellular, ovarian cancer structures derived from serous ovarian cancer cells isolated along disease evolution. It also tests their capacity to cause peritoneal disease in immunosuppressed mice. Results stem from an analysis of cell lines representing the most frequently diagnosed ovarian cancer histotype (high-grade serous ovarian cancer), derived from ascites of the same patient at distinct stages of disease progression. When cultured under adherent conditions, in addition to forming cellular monolayers, the cultures developed areas in which the cells grew upwards, forming densely packed multilayers that ultimately detached from the bottom of the plates and lived as free-floating, multicellular structures. The capacity to form foci and to develop multicellular structures was proportional to disease progression at the time of ascites extraction. Self-assembled in culture, these structures varied in size, were either compact or hollow, irregular, or spheroidal, and exhibited replicative capacity and an epithelial nature. Furthermore, they fully recreated ovarian cancer disease in immunosuppressed mice: accumulation of malignant ascites and pleural effusions; formation of discrete, solid, macroscopic, peritoneal tumors; and microscopic growths in abdominal organs. They also reproduced the histopathological features characteristic of high-grade serous ovarian cancer when diagnosed in patients. The following results encourage the development of therapeutic interventions to interrupt the formation and/or survival of multicellular structures that constitute a floating niche in the peritoneal fluid, which in turn halts disease progression and prevents recurrence.
Collapse
|
10
|
Záveský L, Jandáková E, Weinberger V, Minář L, Hanzíková V, Dušková D, Drábková LZ, Hořínek A. Ovarian Cancer: Differentially Expressed microRNAs in Tumor Tissue and Cell-Free Ascitic Fluid as Potential Novel Biomarkers. Cancer Invest 2019; 37:440-452. [PMID: 31530033 DOI: 10.1080/07357907.2019.1663208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the deadliest gynecologic cancer. The large-scale microRNA (miRNA) expression profiling and individual miRNA validation was performed to find potential novel biomarkers for ovarian cancer. The most consistent overexpression of miRs-200b-3p, 135 b-5p and 182-5p was found in both ascitic fluid and tumors and suggests their potential as oncogenes. miR-451a was consistently underexpressed so may be a tumor suppressor. Results were inconsistent for miR-204-5p, which was overexpressed in ascitic fluid but underexpressed in tumor tissue. miR-203a-3p was generally overexpressed but this failed to be proved in independent sample set in tissue validation.
Collapse
Affiliation(s)
- Luděk Záveský
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| | - Eva Jandáková
- Department of Pathology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Vít Weinberger
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Luboš Minář
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Veronika Hanzíková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | - Daniela Dušková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | | | - Aleš Hořínek
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic.,3rd Department of Medicine, Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| |
Collapse
|
11
|
Silva LM, Kryza T, Stoll T, Hoogland C, Dong Y, Stephens CR, Hastie ML, Magdolen V, Kleifeld O, Gorman JJ, Clements JA. Integration of Two In-depth Quantitative Proteomics Approaches Determines the Kallikrein-related Peptidase 7 (KLK7) Degradome in Ovarian Cancer Cell Secretome. Mol Cell Proteomics 2019; 18:818-836. [PMID: 30705123 DOI: 10.1074/mcp.ra118.001304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Indexed: 12/31/2022] Open
Abstract
Kallikrein-related peptidase 7 (KLK7) is a serine peptidase that is over expressed in ovarian cancer. In vitro functional analyses have suggested KLK7 to play a cancer progressive role, although monitoring of KLK7 expression has suggested a contradictory protective role for KLK7 in ovarian cancer patients. In order to help delineate its mechanism of action and thereby the functional roles, information on its substrate repertoire is crucial. Therefore, in this study a quantitative proteomics approach-PROtein TOpography and Migration Analysis Platform (PROTOMAP)-coupled with SILAC was used for in-depth analysis of putative KLK7 substrates from a representative ovarian cancer cell line, SKOV-3, secreted proteins. The Terminal Amine Isotopic Labeling of Substrates (TAILS) approach was used to determine the exact cleavage sites and to validate qPROTOMAP-identified putative substrates. By employing these two technically divergent approaches, exact cleavage sites on 16 novel putative substrates and two established substrates, matrix metalloprotease (MMP) 2 and insulin growth factor binding protein 3 (IGFBP3), were identified in the SKOV-3 secretome. Eight of these substrates were also identified on TAILS analysis of another ovarian cancer cell (OVMZ-6) secretome, with a further seven OVMZ-6 substrates common to the SKOV-3 qPROTOMAP profile. Identified substrates were significantly associated with the common processes of cell adhesion, extracellular matrix remodeling and cell migration according to the gene ontology (GO) biological process analysis. Biochemical validation supports a role for KLK7 in directly activating pro-MMP10, hydrolysis of IGFBP6 and cleavage of thrombospondin 1 with generation of a potentially bioactive N-terminal fragment. Overall, this study constitutes the most comprehensive analysis of the putative KLK7 degradome in any cancer to date, thereby opening new avenues for KLK7 research.
Collapse
Affiliation(s)
- Lakmali Munasinghage Silva
- From the ‡Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia;; ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany.
| | - Thomas Kryza
- From the ‡Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia;; ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Thomas Stoll
- §Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Christine Hoogland
- §Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia;; ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Ying Dong
- From the ‡Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Carson Ryan Stephens
- From the ‡Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia;; ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Marcus Lachlan Hastie
- §Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Viktor Magdolen
- ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Oded Kleifeld
- ¶Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Victoria, Australia 3800;; ‖Klinische Forschergruppe der Frauenklinik, Klinikum Rechts der Isar, TU München, Munich, Germany
| | - Jeffrey John Gorman
- §Protein Discovery Centre, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Judith Ann Clements
- From the ‡Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia;.
| |
Collapse
|
12
|
Klymenko Y, Wates RB, Weiss-Bilka H, Lombard R, Liu Y, Campbell L, Kim O, Wagner D, Ravosa MJ, Stack MS. Modeling the effect of ascites-induced compression on ovarian cancer multicellular aggregates. Dis Model Mech 2018; 11:dmm034199. [PMID: 30254133 PMCID: PMC6176988 DOI: 10.1242/dmm.034199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/29/2018] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. EOC dissemination is predominantly via direct extension of cells and multicellular aggregates (MCAs) into the peritoneal cavity, which adhere to and induce retraction of peritoneal mesothelium and proliferate in the submesothelial matrix to generate metastatic lesions. Metastasis is facilitated by the accumulation of malignant ascites (500 ml to >2 l), resulting in physical discomfort and abdominal distension, and leading to poor prognosis. Although intraperitoneal fluid pressure is normally subatmospheric, an average intraperitoneal pressure of 30 cmH2O (22.1 mmHg) has been reported in women with EOC. In this study, to enable experimental evaluation of the impact of high intraperitoneal pressure on EOC progression, two new in vitro model systems were developed. Initial experiments evaluated EOC MCAs in pressure vessels connected to an Instron to apply short-term compressive force. A Flexcell Compression Plus system was then used to enable longer-term compression of MCAs in custom-designed hydrogel carriers. Results show changes in the expression of genes related to epithelial-mesenchymal transition as well as altered dispersal of compressed MCAs on collagen gels. These new model systems have utility for future analyses of compression-induced mechanotransduction and the resulting impact on cellular responses related to intraperitoneal metastatic dissemination.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Rebecca B Wates
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Holly Weiss-Bilka
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel Lombard
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Leigh Campbell
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Oleg Kim
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Diane Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew J Ravosa
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - M Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
13
|
Záveský L, Jandáková E, Weinberger V, Minář L, Hanzíková V, Dušková D, Drábková LZ, Svobodová I, Hořínek A. Ascites-Derived Extracellular microRNAs as Potential Biomarkers for Ovarian Cancer. Reprod Sci 2018; 26:510-522. [PMID: 29779470 DOI: 10.1177/1933719118776808] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ovarian cancer as the most fatal gynecological malignancy is often manifested by excessive fluid accumulation known as ascites or effusion. Ascites-derived microRNAs (miRNAs) may be closely associated with ovarian cancer progression. However, our knowledge of their roles, altered expression, and clinical outcomes remained limited. In this study, large-scale expression profiling of 754 human miRNAs was performed using real-time quantitative polymerase chain reaction and 384-well TaqMan array human miRNA A and B cards to identify differentially expressed miRNAs between extracellular fraction of the ascitic fluid associated with high-grade serous ovarian carcinomas and control plasma. Of the 754 miRNAs, 153 were significantly differentially expressed relative to the controls. Expression of 7 individual miRNAs (miR-200a, miR-200b, miR-200c, miR-141, miR-429, miR-1290, and miR-30a-5p) was further validated in extended sample sets, including serous, endometrioid, and mucinous subtypes. All miR-200 family members and miR-1290 were conspicuously overexpressed, while miR-30a-5p was only weakly overexpressed. The ability of miRNAs expression to discriminate the pathological samples from the controls was strong. Receiver operating characteristic curve analyses found area under the curve (AUC) values of 1.000 for miR-200a, miR-200c, miR-141, miR-429, and miR-1290 and of AUC 0.996 and 0.885 for miR-200b and miR-30a-5p, respectively. Preliminary survival analyses indicated low expression level of miR-200b as significantly related to longer overall survival (hazard ratio [HR]: 0.25, mean survival 44 months), while high expression level was related to poor overall survival (HR: 4.04, mean survival 24 months). Our findings suggested that ascites-derived miRNAs should be further explored and evaluated as potential diagnostic and prognostic biomarkers for ovarian cancer.
Collapse
Affiliation(s)
- Luděk Záveský
- 1 First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Eva Jandáková
- 2 Institute of Pathology, University Hospital Brno, Brno, Czech Republic
| | - Vít Weinberger
- 3 Department of Obstetrics and Gynecology, University Hospital Brno and Masaryk University in Brno, Brno, Czech Republic
| | - Luboš Minář
- 3 Department of Obstetrics and Gynecology, University Hospital Brno and Masaryk University in Brno, Brno, Czech Republic
| | - Veronika Hanzíková
- 4 Faculty Transfusion Centre, General University Hospital in Prague, Prague, Czech Republic
| | - Daniela Dušková
- 4 Faculty Transfusion Centre, General University Hospital in Prague, Prague, Czech Republic
| | | | - Iveta Svobodová
- 1 First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Aleš Hořínek
- 1 First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
14
|
Yang L, Zhang X, Ma Y, Zhao X, Li B, Wang H. Ascites promotes cell migration through the repression of miR-125b in ovarian cancer. Oncotarget 2017; 8:51008-51015. [PMID: 28881624 PMCID: PMC5584225 DOI: 10.18632/oncotarget.16846] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/14/2017] [Indexed: 01/04/2023] Open
Abstract
Interactions between ovarian cancer cells and the surrounding tumor microenvironment are not well characterized. Here, we investigated the molecular mechanisms by which malignant ascites promote the metastasis of ovarian cancer. It was found that ovarian cancer ascites promoted ovarian cancer cell migration which was attenuated by either heat inactivation or antibody blockade of TGF-β. High level (at ng/ml level) of TGF-β was detected in the ascites. In addition, ascites repressed the expression of miRNA-125b in a TGF-β-dependent manner. Mimic of miR-125b blocked ascites-induced cell migration. Furthermore, Gab2 (a target gene of miR-125b) was elevated by ascites in a TGF-β-dependent manner. And forced expression of Gab2 reversed the inhibition of migration induced by miR-125b mimic. Most importantly, the expression of miR-125b and Gab2 mRNA was negatively correlated in ovarian cancer specimens. Taken together, our finding suggested that TGF-β in ascites promoted cancer cell migration through repression of miR-125b in ovarian cancer. This might provide a novel therapeutic target for ovarian cancer in the future.
Collapse
Affiliation(s)
- Lan Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Bin Li
- Department of Gynecological Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| |
Collapse
|
15
|
Nawaz M, Fatima F, Nazarenko I, Ekström K, Murtaza I, Anees M, Sultan A, Neder L, Camussi G, Valadi H, Squire JA, Kislinger T. Extracellular vesicles in ovarian cancer: applications to tumor biology, immunotherapy and biomarker discovery. Expert Rev Proteomics 2016; 13:395-409. [PMID: 26973172 DOI: 10.1586/14789450.2016.1165613] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years there has been tremendous interest in both the basic biology and applications of extracellular vesicles (EVs) in translational cancer research. This includes a better understanding of their biogenesis and mechanisms of selective cargo packaging, their precise roles in horizontal communication, and their application as non-invasive biomarkers. The rapid advances in next-generation omics technologies are the driving forces for these discoveries. In this review, the authors focus on recent results of EV research in ovarian cancer. A deeper understanding of ovarian cancer-derived EVs, the types of cargo molecules and their biological roles in cancer growth, metastases and drug resistance, could have significant impact on the discovery of novel biomarkers and innovative therapeutics. Insights into the role of EVs in immune regulation could lead to novel approaches built on EV-based immunotherapy.
Collapse
Affiliation(s)
- Muhammad Nawaz
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Farah Fatima
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Irina Nazarenko
- c Institute for Environmental Health Sciences and Hospital Infection Control , University Medical Centre Freiburg , Freiburg im Breisgau , Germany
| | - Karin Ekström
- d Department of Biomaterials , Sahlgrenska Academy at the University of Gothenburg , Gothenburg , Sweden.,e BIOMATCELL VINN Excellence Centre of Biomaterials and Cell Therapy , Gothenburg , Sweden
| | - Iram Murtaza
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Mariam Anees
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Aneesa Sultan
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Luciano Neder
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Giovanni Camussi
- g Department of Medical Sciences and Molecular Biotechnology Centre , University of Torino , Torino , Italy
| | - Hadi Valadi
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Jeremy A Squire
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Thomas Kislinger
- h Princess Margaret Cancer Centre and Department of Medical Biophysics , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
16
|
Monk BJ, Poveda A, Vergote I, Raspagliesi F, Fujiwara K, Bae DS, Oaknin A, Ray-Coquard I, Provencher DM, Karlan BY, Lhommé C, Richardson G, Rincón DG, Coleman RL, Marth C, Brize A, Fabbro M, Redondo A, Bamias A, Ma H, Vogl FD, Bach BA, Oza AM. Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2. Gynecol Oncol 2016; 143:27-34. [PMID: 27546885 DOI: 10.1016/j.ygyno.2016.07.112] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/12/2016] [Accepted: 07/23/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Trebananib, a peptibody that blocks binding of angiopoietin-1 and -2 to Tie2, significantly prolonged progression-free survival (PFS) in patients with recurrent epithelial ovarian cancer in the phase 3 TRINOVA-1 study. We report overall survival (OS) in the intent-to-treat population and clinically relevant subgroups and time to second disease progression (PFS-2). PATIENTS AND METHODS Women with recurrent disease (platinum-free interval<12months) were randomized to receive intravenous paclitaxel 80mg/m(2) (3weeks on/1week off) plus intravenous trebananib 15mg/kg or placebo, weekly. OS in the intent-to-treat population was a key secondary endpoint. Exploratory analysis of PFS-2 was conducted according to guidance by the European Medicines Agency. RESULTS Median OS was not significantly improved with trebananib compared with placebo (19.3 versus 18.3months; HR, 0.95; 95% CI, 0.81-1.11; P=0.52) in the intent-to-treat population (n=919). In subgroup analysis, trebananib improved median OS compared with placebo (14.5 versus 12.3months; HR, 0.72; 95% CI, 0.55-0.93; P=0.011) in patients with ascites at baseline (n=295). In the intent-to-treat population, trebananib significantly improved median PFS-2 compared with placebo (12.5 versus 10.9months; HR, 0.85; 95% CI, 0.74-0.98; P=0.024). The incidence and type of adverse events in this updated analysis was consistent with that described in the primary analysis; no new safety signals were detected. CONCLUSIONS OS was not significantly longer in the intent-to-treat population, although there was an improvement in OS in patients with ascites receiving trebananib. PFS-2 confirmed that the PFS benefit associated with trebananib was maintained through the second disease progression independent of the choice of subsequent therapy.
Collapse
Affiliation(s)
- Bradley J Monk
- Department of Obstetrics and Gynecology, University of Arizona Cancer Center at Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - Andrés Poveda
- Area Clinica de Oncologia Ginecológica, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Ignace Vergote
- Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven Cancer Institute, KU Leuven, European Union, Belgium
| | - Francesco Raspagliesi
- Gynecologic Oncology Unit, Fondazione IRCCS, Istituto Nazionale per la Cura e lo Studio dei Tumori, Milano, Italy
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka-Shi, Japan
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, South Korea
| | - Ana Oaknin
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Diane M Provencher
- Division of Gynecologic Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Catherine Lhommé
- Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Gary Richardson
- Academic Haematology and Oncology, Cabrini Hospital, Malvern, VIC, Australia
| | | | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christian Marth
- Universitätsklinik für Gynäkologie und Geburtshilfe, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Arija Brize
- Latvian Oncology Center, Riga Eastern Clinical University Hospital, Riga, Latvia
| | - Michel Fabbro
- Regional Cancer Institute Montpellier, Montpellier, France
| | | | - Aristotelis Bamias
- Alexandra Hospital, Department of Clinical Therapeutics, National & Kapodistrian University of Athens, Athens, Greece
| | - Haijun Ma
- Global Biostatistical Science, Amgen Inc., Thousand Oaks, CA, USA
| | - Florian D Vogl
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Bruce A Bach
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Amit M Oza
- Department of Medicine, Princess Margaret Hospital, University of Toronto, ON, Canada
| |
Collapse
|
17
|
Thomsen J, Hjortebjerg R, Espelund U, Ørtoft G, Vestergaard P, Magnusson NE, Conover CA, Tramm T, Hager H, Høgdall C, Høgdall E, Oxvig C, Frystyk J. PAPP-A proteolytic activity enhances IGF bioactivity in ascites from women with ovarian carcinoma. Oncotarget 2016; 6:32266-78. [PMID: 26336825 PMCID: PMC4741676 DOI: 10.18632/oncotarget.5010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 08/14/2015] [Indexed: 11/25/2022] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) stimulates insulin-like growth factor (IGF) action through proteolysis of IGF-binding protein (IGFBP)-4. In experimental animals, PAPP-A accelerates ovarian tumor growth by this mechanism. To investigate the effect of PAPP-A in humans, we compared serum and ascites from 22 women with ovarian carcinoma. We found that ascites contained 46-fold higher PAPP-A levels as compared to serum (P < 0.001). The majority (80%) of PAPP-A was enzymatically active. This is supported by the finding that ascites contained more cleaved than intact IGFBP-4 (P < 0.03). Ascites was more potent than serum in activating the IGF-I receptor (IGF-IR) in vitro (+31%, P < 0.05); in 8 of 22 patients by more than two-fold. In contrast, ascites contained similar levels of immunoreactive IGF-I, and lower levels of IGF-II (P < 0.001). Immunohistochemistry demonstrated the presence of IGF-IR in all but one tumor, whereas all tumors expressed PAPP-A, IGFBP-4, IGF-I and IGF-II. Addition of recombinant PAPP-A to ascites increased the cleavage of IGFBP-4 and enhanced IGF-IR activation (P < 0.05). In conclusion, human ovarian tumors express PAPP-A, IGFBP-4 and IGFs and these proteins are also present in ascites. We suggest that both soluble PAPP-A in ascites and tissue-associated PAPP-A serve to increase IGF bioactivity and, thereby, to stimulate IGF-IR-mediated tumor growth.
Collapse
Affiliation(s)
- Jacob Thomsen
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Ulrick Espelund
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Gitte Ørtoft
- Department of Gynecology, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Poul Vestergaard
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | - Nils E Magnusson
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Cheryl A Conover
- Division of Endocrinology and Metabolism, Endocrine Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | - Henrik Hager
- Department of Pathology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | - Claus Høgdall
- Clinic of Gynecology, Juliane Marie Centret, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Faculty of Science & Technology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| |
Collapse
|
18
|
Meyer L, Suidan R, Sun C, Westin S, Coleman RL, Mills GB. The management of malignant ascites and impact on quality of life outcomes in women with ovarian cancer. EXPERT REVIEW OF QUALITY OF LIFE IN CANCER CARE 2016; 1:231-238. [PMID: 30906877 PMCID: PMC6425954 DOI: 10.1080/23809000.2016.1185369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant ascites is one of the most common sequela of epithelial ovarian cancer. It causes significant symptoms and can have a detrimental impact on patient quality of life, especially in women with recurrent ovarian cancer. The management of symptomatic ascites consists of both mechanical treatments that aim to drain the peritoneal cavity, and medical therapies that prevent and diminish the development of ascites. Mechanical options include serial paracentesis, peritoneal catheters, and peritoneovenous shunts. Pharmaceutical treatments include diuretics, angiogenesis inhibitors, and other targeted agents. There is a perception, without formal analysis, that intractable ascites is less common in the taxane era of therapy. In this review paper, we highlight current and emerging therapeutic strategies, complications and contraindications, and their effects on patient quality of life.
Collapse
Affiliation(s)
- Larissa Meyer
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Rudy Suidan
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Charlotte Sun
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Shannon Westin
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Robert L Coleman
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Gordon B Mills
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| |
Collapse
|
19
|
Bobbs AS, Cole JM, Cowden Dahl KD. Emerging and Evolving Ovarian Cancer Animal Models. CANCER GROWTH AND METASTASIS 2015; 8:29-36. [PMID: 26380555 PMCID: PMC4558890 DOI: 10.4137/cgm.s21221] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 12/23/2022]
Abstract
Ovarian cancer (OC) is the leading cause of death from a gynecological malignancy in the United States. By the time a woman is diagnosed with OC, the tumor has usually metastasized. Mouse models that are used to recapitulate different aspects of human OC have been evolving for nearly 40 years. Xenograft studies in immunocompromised and immunocompetent mice have enhanced our knowledge of metastasis and immune cell involvement in cancer. Patient-derived xenografts (PDXs) can accurately reflect metastasis, response to therapy, and diverse genetics found in patients. Additionally, multiple genetically engineered mouse models have increased our understanding of possible tissues of origin for OC and what role individual mutations play in establishing ovarian tumors. Many of these models are used to test novel therapeutics. As no single model perfectly copies the human disease, we can use a variety of OC animal models in hypothesis testing that will lead to novel treatment options. The goal of this review is to provide an overview of the utility of different mouse models in the study of OC and their suitability for cancer research.
Collapse
Affiliation(s)
- Alexander S Bobbs
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN, USA. ; Harper Cancer Research Institute, South Bend, IN, USA
| | - Jennifer M Cole
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN, USA. ; Harper Cancer Research Institute, South Bend, IN, USA
| | - Karen D Cowden Dahl
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN, USA. ; Harper Cancer Research Institute, South Bend, IN, USA. ; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA. ; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
20
|
Stommel MWJ, Strik C, van Goor H. Response to pathological processes in the peritoneal cavity--sepsis, tumours, adhesions, and ascites. Semin Pediatr Surg 2014; 23:331-5. [PMID: 25459437 DOI: 10.1053/j.sempedsurg.2014.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The peritoneum is one of the commonest sites for pathological processes in pediatric surgery. Its response to pathological processes is characterized by an inflammatory reaction with specific pathways depending on the type of injury or peritoneal process involved. This review discusses the current understanding of peritoneal inflammation, adhesion formation, intra-abdominal sepsis, peritoneal metastasis, and ascites and briefly reviews new therapeutic strategies to treat or prevent these pathological entities. Recent studies have improved the understanding of peritoneal responses, resulting in possible new targets for prevention and therapy.
Collapse
Affiliation(s)
- Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Chema Strik
- Department of Surgery, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Harry van Goor
- Department of Surgery, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
21
|
Ezzati M, Abdullah A, Shariftabrizi A, Hou J, Kopf M, Stedman JK, Samuelson R, Shahabi S. Recent Advancements in Prognostic Factors of Epithelial Ovarian Carcinoma. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:953509. [PMID: 27382614 PMCID: PMC4897239 DOI: 10.1155/2014/953509] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/26/2014] [Indexed: 01/03/2023]
Abstract
Ovarian cancer remains the most common cause of gynecologic cancer-related death among women in developed countries. Nevertheless, subgroups of ovarian cancer patients experience relatively longer survival. Efforts to identify prognostic factors that characterize such patients are ongoing, with investigational areas including tumor characteristics, surgical management, inheritance patterns, immunologic factors, and genomic patterns. This review discusses various demographic, clinical, and molecular factors implicating longevity and ovarian cancer survival. Continued efforts at identifying these prognosticators may result in invaluable adjuncts to the treatment of ovarian cancer, with the ultimate goal of advancing patient care.
Collapse
Affiliation(s)
- Mohammad Ezzati
- Department of Obstetrics and Gynecology, Washington Hospital Center, 110 Irving Street NW, Washington, DC 20010, USA
- Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Amer Abdullah
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Ahmad Shariftabrizi
- Department of Pathology and Laboratory Medicine, School of Medicine, Tufts University, 800 Washington Street, Boston, MA 02111, USA
| | - June Hou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine and Montefiore Medical Center, Montefiore Centennial, 3332 Rochambeau Avenue, Bronx, NY 10467-2836, USA
| | - Michael Kopf
- Department of Medicine, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Jennifer K. Stedman
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Robert Samuelson
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Shohreh Shahabi
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| |
Collapse
|
22
|
Anuradha S, Webb PM, Blomfield P, Brand AH, Friedlander M, Leung Y, Obermair A, Oehler MK, Quinn M, Steer C, Jordan SJ. Survival of Australian women with invasive epithelial ovarian cancer: a population‐based study. Med J Aust 2014; 201:283-8. [DOI: 10.5694/mja14.00132] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/27/2014] [Indexed: 11/17/2022]
Affiliation(s)
- Satyamurthy Anuradha
- Gynaecological Cancers Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Penelope M Webb
- Gynaecological Cancers Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | | | - Alison H Brand
- Department of Gynaecological Oncology, Westmead Hospital, University of Sydney, Sydney, NSW
| | | | - Yee Leung
- School of Women's and Infants' Health, University of Western Australia, Perth, WA
| | - Andreas Obermair
- Department of Gynaecological Oncology, Royal Brisbane and Women's Hospital, Brisbane, QLD
| | - Martin K Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA
| | - Michael Quinn
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC
| | | | - Susan J Jordan
- Gynaecological Cancers Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| |
Collapse
|
23
|
Dong Y, Loessner D, Irving-Rodgers H, Obermair A, Nicklin JL, Clements JA. Metastasis of ovarian cancer is mediated by kallikrein related peptidases. Clin Exp Metastasis 2014; 31:135-47. [PMID: 24043563 PMCID: PMC3892111 DOI: 10.1007/s10585-013-9615-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022]
Abstract
Ovarian cancer, in particular epithelial ovarian cancer (EOC), is commonly diagnosed when the tumor has metastasized into the abdominal cavity with an accumulation of ascites fluid. Combining histopathology and genetic variations, EOC can be sub-grouped into Type-I and Type-II tumors, of which the latter are more aggressive and metastatic. Metastasis and chemoresistance are the key events associated with the tumor microenvironment that lead to a poor patient outcome. Kallikrein-related peptidases (KLKs) are aberrantly expressed in EOC, in particular, in the more metastatic Type-II tumors. KLKs are a family of 15 serine proteases that are expressed in diverse human tissues and involved in various patho-physiological processes. As extracellular enzymes, KLKs function in the hydrolysis of growth factors, proteases, cell membrane bound receptors, adhesion proteins, and cytokines initiating intracellular signaling pathways and their downstream events. High KLK levels are differentially associated with the prognosis of ovarian cancer patients, suggesting that they not only have application as biomarkers but also function in disease progression, and therefore are potential therapeutic targets. Recent studies have demonstrated the function of these proteases in promoting and/or suppressing the invasive behavior of ovarian cancer cells in metastasis in vitro and in vivo. Both conventional cell culture methods and three-dimensional platforms have been applied to mimic the ovarian cancer microenvironment of patients, such as the solid stromal matrix and ascites fluid. Here we summarize published studies to provide an overview of our understanding of the role of KLKs in EOC, and to lay the foundation for future research directions.
Collapse
Affiliation(s)
- Ying Dong
- Cancer Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia,
| | | | | | | | | | | |
Collapse
|
24
|
Takal MK, Baykal C, Başer E, Kaya MD, Dursun P, Ozen O, Haberal AN, Ayhan A. Does Aquaporin-1 expression have clinical significance in serous epithelial ovarian cancer? J Turk Ger Gynecol Assoc 2013; 14:130-5. [PMID: 24592091 DOI: 10.5152/jtgga.2013.02679] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/16/2013] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE To assess the relationship between Aquaporin-1 (AQP1) expression and clinicopathological variables in serous epithelial ovarian cancer (EOC). MATERIAL AND METHODS Serous EOC cases treated in our institution between January 2007 and December 2009 were included in the study. A semi-quantitative immunohistochemical method was used to determine AQP1 expression levels, intratumoral microvessel density (IMD) and AQP1/IMD ratios. The relationship between these parameters and clinicopathological variables were assessed. P values less than 0.05 was considered statistically significant. RESULTS A total of 55 cases of serous EOC were included in the study. AQP1 was strongly expressed in the membranes of microvessels and small vessels within all tumor tissues. In a few cases, AQP1 expression was also observed in the membrane of interstitial cells and in individual tumor cells. A positive correlation was detected between preoperative CA125 levels and the expression of AQP1 (R: 0.277, p<0.05). AQP1 expression was similar between FIGO stage I-II and FIGO stage III-IV cases (p > 0.05). A significant relationship did not exist between AQP1 expression and FIGO stage, lymph node metastasis or ascites volume (p>0.05). CONCLUSION In this study, AQP1 expression did not have a significant association with important clinicopathological variables in serous EOC. Future studies are needed to determine AQP1 expression in other histological types of EOC.
Collapse
Affiliation(s)
- Mustafa Kemal Takal
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| | - Cem Baykal
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| | - Eralp Başer
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| | - Mustafa Derda Kaya
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| | - Polat Dursun
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| | - Ozlem Ozen
- Department of Pathology, Başkent University School of Medicine, Ankara, Turkey
| | | | - Ali Ayhan
- Department of Obstetrics and Gynecology, Division of Gynecological Oncology, Başkent University School of Medicine, Ankara, Turkey
| |
Collapse
|
25
|
Dong Y, Stephens C, Walpole C, Swedberg JE, Boyle GM, Parsons PG, McGuckin MA, Harris JM, Clements JA. Paclitaxel resistance and multicellular spheroid formation are induced by kallikrein-related peptidase 4 in serous ovarian cancer cells in an ascites mimicking microenvironment. PLoS One 2013; 8:e57056. [PMID: 23451143 PMCID: PMC3581584 DOI: 10.1371/journal.pone.0057056] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 01/16/2013] [Indexed: 12/14/2022] Open
Abstract
High tumor kallikrein-related-peptidase 4 (KLK4) levels are associated with a poor outcome for women with serous epithelial ovarian cancer (EOC), for which peritoneal dissemination and chemoresistance are key events. To determine the role of KLK4 in these events, we examined KLK4-transfected SKOV-3 and endogenous KLK4 expressing OVCA432 cells in 3-dimensional (3D) suspension culture to mimic the ascites microenvironment. KLK4-SKOV-3 cells formed multicellular aggregates (MCAs) as seen in ascites, as did SKOV-3 cells treated with active KLK4. MCA formation was reduced by treatment with a KLK4 blocking antibody or the selective active site KLK4 sunflower trypsin inhibitor (SFTI-FCQR). KLK4-MCAs formed larger cancer cell foci in mesothelial cell monolayers than those formed by vector and native SKOV-3 cells, suggesting KLK4-MCAs are highly invasive in the peritoneal microenvironment. A high level of KLK4 is expressed by ascitic EOC cells compared to matched primary tumor cells, further supporting its role in the ascitic microenvironment. Interestingly, KLK4 transfected SKOV-3 cells expressed high levels of the KLK4 substrate, urokinase plasminogen activator (uPA), particularly in 3D-suspension, and high levels of both KLK4 and uPA were observed in patient cells taken from ascites. Importantly, the KLK4-MCAs were paclitaxel resistant which was reversed by SFTI-FCQR and to a lesser degree by the general serine protease inhibitor, Aprotinin, suggesting that in addition to uPA, other as yet unidentified substrates of KLK4 must be involved. Nonetheless, these data suggest that KLK4 inhibition, in conjunction with paclitaxel, may improve the outcome for women with serous epithelial ovarian cancer and high KLK4 levels in their tumors.
Collapse
Affiliation(s)
- Ying Dong
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Carson Stephens
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Carina Walpole
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Joakim E. Swedberg
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Glen M. Boyle
- Drug Discovery Group, Division of Cancer and Cell Biology, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Peter G. Parsons
- Drug Discovery Group, Division of Cancer and Cell Biology, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Michael A. McGuckin
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - Jonathan M. Harris
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Judith A. Clements
- Cancer Program, Institute of Health and Biomedical Innovation and Faculty of Sciences and Technology, Queensland University of Technology, Kelvin Grove, Queensland, Australia
- * E-mail:
| |
Collapse
|
26
|
Abstract
The peritoneal metastatic route of cancer dissemination is shared by cancers of the ovary and gastrointestinal tract. Once initiated, peritoneal metastasis typically proceeds rapidly in a feed-forward manner. Several factors contribute to this efficient progression. In peritoneal metastasis, cancer cells exfoliate into the peritoneal fluid and spread locally, transported by peritoneal fluid. Inflammatory cytokines released by tumor and immune cells compromise the protective, anti-adhesive mesothelial cell layer that lines the peritoneal cavity, exposing the underlying extracellular matrix to which cancer cells readily attach. The peritoneum is further rendered receptive to metastatic implantation and growth by myofibroblastic cell behaviors also stimulated by inflammatory cytokines. Individual cancer cells suspended in peritoneal fluid can aggregate to form multicellular spheroids. This cellular arrangement imparts resistance to anoikis, apoptosis, and chemotherapeutics. Emerging evidence indicates that compact spheroid formation is preferentially accomplished by cancer cells with high invasive capacity and contractile behaviors. This review focuses on the pathological alterations to the peritoneum and the properties of cancer cells that in combination drive peritoneal metastasis.
Collapse
|
27
|
Decruze SB, Macdonald R, Smith G, Herod JJ. Paracentesis in ovarian cancer: a study of the physiology during free drainage of ascites. J Palliat Med 2010; 13:251-4. [PMID: 19929225 DOI: 10.1089/jpm.2009.0158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ascites is frequently seen in patients suffering with ovarian cancer. Paracentesis is a well-established method to provide symptomatic relief. Systemic adverse effects have occurred in paracentesis for ascites arising from other conditions. However, malignancy-related ascites has a different pathophysiology than nonmalignant ascites and as such the concerns related to the latter condition may not apply in patients with ovarian cancer. This study has shown that paracentesis is both effective and safe as an outpatient procedure for managing malignancy-related ascites.
Collapse
Affiliation(s)
- Shandya Bridget Decruze
- Department of Gynaecological Oncology, Liverpool Womens NHS Foundation Trust, Liverpool, United Kingdom.
| | | | | | | |
Collapse
|
28
|
Lane D, Goncharenko-Khaider N, Rancourt C, Piché A. Ovarian cancer ascites protects from TRAIL-induced cell death through alphavbeta5 integrin-mediated focal adhesion kinase and Akt activation. Oncogene 2010; 29:3519-31. [PMID: 20400979 DOI: 10.1038/onc.2010.107] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Interactions between ovarian cancer cells and the surrounding tumor microenvironment are not well characterized. We have earlier shown that ovarian cancer ascites induces Akt activation and protect tumor cells from TRAIL-induced apoptosis. Here, we investigated the mechanism by which ascites activates Akt. The ability of ovarian cancer ascites to activate Akt and inhibit TRAIL-induced cell death and caspase activity was decreased by heat inactivation, but was retained in ascites fractions >5 kDa. The survival promoting activity of ascites was not affected by inhibitors of growth factor receptor including epidermal growth factor receptor (EGFR), VEGFR, FGFR, Her2/neu, and IGF-R1. However, this activity was inhibited by an alphavbeta5 integrin-blocking antibody, but not by blocking antibodies against alphavbeta3, beta1, or beta3 integrins. alphavbeta5 integrin-blocking antibodies also inhibited ascites-induced Akt phosphorylation and c-FLIPs up-regulation. Ovarian cancer ascites induced a rapid phosphorylation of focal adhesion kinase (FAK), which closely correlated with the phosphorylation of Akt overtime. FAK phosphorylation was strongly inhibited by alphavbeta5 integrin-blocking antibodies. Depletion of FAK content by RNA interference was also associated with inhibition of ascites-mediated Akt activation and survival. These results suggest that ovarian cancer ascites induces FAK and Akt activation in an alphavbeta5 integrin-dependent pathway, which confers protection from TRAIL-induced cell death and caspase activation.
Collapse
Affiliation(s)
- D Lane
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | | | | | | |
Collapse
|
29
|
|
30
|
Sodek KL, Ringuette MJ, Brown TJ. Compact spheroid formation by ovarian cancer cells is associated with contractile behavior and an invasive phenotype. Int J Cancer 2009; 124:2060-70. [PMID: 19132753 DOI: 10.1002/ijc.24188] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ovarian cancer cells are present in malignant ascites both as individual cells and as multicellular spheroid aggregates. Although spheroid formation affords protection of cancer cells against some chemotherapeutic agents, it has not been established whether a relationship exists between invasive behavior and predisposition to spheroid formation. Aspects of spheroid formation, including cell-matrix adhesion, remodeling and contractility are characteristic myofibroblast-like behaviors associated with fibrosis that contribute to tumor growth and dissemination. We explored the possibility that cell behaviors that promote spheroid formation also facilitate invasion. Our analysis of 6 human ovarian cancer cell lines indicated that ovarian cancer cells possessing myofibroblast-like properties formed compact spheroids and invaded 3D matrices. These cells readily contracted collagen I gels, possessed a spindle-like morphology, and had elevated expression of genes associated with the TGFbeta-mediated fibrotic response and/or beta1 integrin function, including fibronectin (FN), connective tissue growth factor (CTGF/CCN2), lysyl oxidase (LOX1), tissue transglutaminase 2 (TGM2) and urinary plasminogen activator receptor (uPAR). Whereas cell aggregation was induced by TGFbeta, and by beta1-integrin overexpression and activation, these treatments did not stimulate the contractile activity required for spheroid compaction. The positive relationship found between compact spheroid formation and invasive behavior implies a preferential survival of an invasive subpopulation of ovarian cancer cells, as cells in spheroids are more resistant to several chemotherapeutics. Preventing the formation of ovarian cancer spheroids may represent a novel strategy to improve the efficacy of existing therapeutics.
Collapse
Affiliation(s)
- Katharine L Sodek
- Department of Cell and Systems Biology, University of Toronto, Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | | | | |
Collapse
|
31
|
Bak SP, Walters JJ, Takeya M, Conejo-Garcia JR, Berwin BL. Scavenger receptor-A-targeted leukocyte depletion inhibits peritoneal ovarian tumor progression. Cancer Res 2007; 67:4783-9. [PMID: 17510407 DOI: 10.1158/0008-5472.can-06-4410] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunosuppressive leukocytes are emerging as a critical factor in facilitating tumor progression. These leukocytes are converted by the tumor microenvironment to become tolerogenic, facilitate metastasis, and to aid in neovascularization. The predominant variety of suppressive leukocytes found in human and murine ovarian cancer are called vascular leukocytes (VLC), due to sharing functions and cell surface markers of both dendritic cells and endothelial cells. Using the ID8 murine model of ovarian cancer, the aim of this study was to test the efficacy of VLC elimination as an ovarian tumor therapy. We show that carrageenan-mediated depletion of peritoneal tumor-associated leukocytes inhibits ovarian tumor progression. We then identified scavenger receptor-A (SR-A) as a cell surface receptor that is robustly and specifically expressed within human and murine ovarian tumor ascites upon VLCs. Administration of anti-SR-A immunotoxin to mice challenged with peritoneal ID8 tumors eliminated tumor-associated VLCs and, importantly, substantially inhibited peritoneal tumor burden and ascites accumulation. Moreover, the toxin required targeting to SR-A because mice that received untargeted toxin did not exhibit inhibition of tumor progression. We conclude that SR-A constitutes a novel and specific target for efficacious immunotherapeutic treatment of peritoneal ovarian cancer.
Collapse
Affiliation(s)
- S Peter Bak
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA
| | | | | | | | | |
Collapse
|
32
|
Ayhan A, Gultekin M, Taskiran C, Dursun P, Firat P, Bozdag G, Celik NY, Yuce K. Ascites and epithelial ovarian cancers: a reappraisal with respect to different aspects. Int J Gynecol Cancer 2007; 17:68-75. [PMID: 17291234 DOI: 10.1111/j.1525-1438.2006.00777.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Ascites is a common finding in patients with epithelial ovarian cancer (EOC). Clinico-pathologic correlations with respect to the presence of ascites, positive cytology and prognostic role of ascites, and the impact of ascitic volumes were not previously studied extensively. A total of 372 patients with EOC were retrospectively evaluated with respect to presence and amount of ascites, cytologic findings, and survival. Two groups were compared by using Chi-square, Student's t and Mann-Whitney U, binary logistic regression, Kaplan Meier and Cox-regression analysis tests, where appropriate. Omental metastasis (P < 0.001; OR: 3.21, 95% CI = 1.945-5.297) and mean number of metastatic lymph nodes (P= 0.008; OR: 1.063, 95% CI = 1.016-1.112) were significantly related with presence of ascites. Evaluation of ascitic volume at different thresholds revealed lymphatic-omental metastasis, and also the disease stage to be significantly different among patient groups at lower threshold values and the positive cytology and high-grade diseases at higher threshold values. In conclusion, presence of ascites correlates with both the intraperitoneal and also the retroperitoneal tumor spread. Amount of ascites has different correlations with the clinico-pathologic factors depending on the thresholds chosen. At lower volumes, lymphatic and omental metastasis seems to correlate with the development of ascites. Once ascites develops, tumor grade seems to be important for larger ascites volumes. Neither the presence of ascites or its volume nor the cytologic positivity was an independent predictor of survival.
Collapse
Affiliation(s)
- A Ayhan
- Departments of Obstetrics and Gynecology and Pathology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Davidson B, Espina V, Steinberg SM, Flørenes VA, Liotta LA, Kristensen GB, Tropé CG, Berner A, Kohn EC. Proteomic Analysis of Malignant Ovarian Cancer Effusions as a Tool for Biologic and Prognostic Profiling. Clin Cancer Res 2006; 12:791-9. [PMID: 16467090 DOI: 10.1158/1078-0432.ccr-05-2516] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Malignant epithelial ovarian cancer effusions are important in disease dissemination and clinical outcome. The identification of biochemical events active in effusions may improve our identification and application of targeted therapeutics. EXPERIMENTAL DESIGN Archival effusion samples for which outcome information was known were studied. Clinical variables were comparable between these groups. Two cohorts of patients with malignant effusion were assessed: those with effusion at presentation (Tap1) or at first recurrence (Tap2). Expression and activated fraction of selected signaling proteins were quantitated on serial protein microarrays using validated antibodies. Proteomic results and clinical variables were analyzed by univariate analysis followed by Cox proportional hazards model analysis. RESULTS Malignant effusions (>80% malignant cells) were distinguished from benign effusions by higher expression of AKT, activated extracellular signal-regulated kinase, activated (P < or = 0.001) and total cAMP-responsive element binding protein (P = 0.01), and JNK (P = 0.03). Malignant pleural effusions could not be differentiated from ascites by signaling profiles. Both had signal expression clusters for survival, proliferation and metastasis, and injury pathways. Cox proportional hazards model analysis revealed high p38 and pEGFR/EGFR ratio as jointly associated with poor survival in Tap1 cases (both P < or = 0.002). Phospho-JNK quantity was associated with worse outcome in Tap2 patients (P = 0.004), when taking other factors into consideration. CONCLUSIONS Proliferation, survival, and apoptosis signaling dysregulation can be identified in ovarian cancer effusion samples. Biochemical characterization of clinical effusions may provide either predictive and/or correlative information on patient outcome from which to further understand the mechanisms of effusion development and target clinical intervention.
Collapse
Affiliation(s)
- Ben Davidson
- Molecular Signaling Section, Laboratory of Pathology, National Cancer Institute, Bethesda Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
von Gruenigen VE, Daly BJ. Futility: clinical decisions at the end-of-life in women with ovarian cancer. Gynecol Oncol 2005; 97:638-44. [PMID: 15863171 DOI: 10.1016/j.ygyno.2005.01.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Revised: 01/24/2005] [Accepted: 01/24/2005] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The purpose of this article is to provide a review of the clinical meaning of futility, discuss current normative uses of futility assessments and propose guidelines for clinicians to use in dialogue regarding treatment decisions for patients with advanced ovarian cancers. METHODS We performed a MEDLINE literature search of relevant clinical articles for this review that discussed futility and the application to women with ovarian cancer. RESULTS Medical futility refers to treatments that serve no physiologic, quantitative or qualitative meaningful purpose. Despite the growth in options focused on symptom management rather than disease eradication, including hospice programs and the more recent development of palliative care programs, there is evidence that many patients continue to receive aggressive interventions, including chemotherapy, until days before their death. While the legal and moral acceptability of treatment limitation is well established, clarity in establishing goals of care, timing of the transition from cure to palliation and communication of specific decisions to withhold further aggressive interventions remain problematic for both patients and clinicians. CONCLUSIONS There continues to be a distinct need for both better understanding of the dynamics of patient choice and increased education of physicians in addressing end-of-life care planning. It is essential that we continue to test specific communication and supportive interventions that will improve our ability to help patients avoid the burden of futile therapy while maintaining hope.
Collapse
Affiliation(s)
- Vivian E von Gruenigen
- Department of Reproductive Biology, Division of Gynecologic Oncology, University MacDonald Women's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | | |
Collapse
|
35
|
Recombinant Adeno-Associated Virus 2-Mediated Antiangiogenic Prevention in a Mouse Model of Intraperitoneal Ovarian Cancer. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.1342.11.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: In the present study, we sought to determine the potential of sustained transgene expression by a single i.m. administration of recombinant adeno-associated virus 2 (rAAV) encoding angiostatin and endostatin in inhibiting i.p. ovarian cancer growth and dissemination in a preclinical mouse model.
Experimental Design: Cohorts of female athymic nude mice received either no virus or 1.2 × 1011 particles of rAAV encoding green fluorescence protein or endostatin plus angiostatin, i.m. Three weeks later, the mice were i.p. injected with 106 human epithelial ovarian cancer cell line SKOV3.ip1. As a measure of effectiveness of the therapy, tumor weight, abdominal distension, ascites volume and vascular endothelial growth factor level, and tumor weight were determined. Immunohistochemistry was done to determine tumor cell apoptosis and endothelial cell proliferation following the therapy. Tumor-free survival was recorded as the end point.
Results: Results indicated a significant tumor-free survival (P < 0.003) following therapy with rAAV encoding endostatin and angiostatin compared with untreated or rAAV-green fluorescence protein–treated mice. Ascites volume in rAAV endostatin and angiostatin–treated mice was significantly lower than naive mice and contained less hemorrhage and tumor conglomerates. The level of vascular endothelial growth factor in the ascites of antiangiogenic vector treated mice was also significantly less compared with the untreated mice. Immunohistochemical analyses indicated increased tumor cell apoptosis and decreased blood vasculature following rAAV endostatin and angiostatin treatment.
Conclusion: The results indicate that antiangiogenic genetic prevention from stable systemic levels of angiostatin and endostatin by i.m. administration of rAAV can be used for the treatment of i.p. ovarian cancer growth and dissemination.
Collapse
|
36
|
Graves LE, Ariztia EV, Navari JR, Matzel HJ, Stack MS, Fishman DA. Proinvasive Properties of Ovarian Cancer Ascites-Derived Membrane Vesicles. Cancer Res 2004; 64:7045-9. [PMID: 15466198 DOI: 10.1158/0008-5472.can-04-1800] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Malignant ovarian ascites are rich in cellular components, membrane-bound vesicles, and soluble proteins. This study focused on the structure of membrane-bound vesicles and their ability to promote invasion in cultured malignant ovarian epithelium. Membrane vesicles were derived from women with stage I-IV malignant ovarian ascites and from nonmalignant gynecologic ascites. Isolated vesicles were characterized by immunofluorescence and Western blot analysis. Using gel zymography for matrix metalloproteinase (MMP) detection and a colorimetric assay for urokinase-type plasminogen activator (uPA) analysis, we analyzed the proteinase activities of MMP-2, MMP-9, and uPA from the prepared vesicles, whole cells isolated from ascites, and the cell-free ultracentrifuged supernatant. The invasiveness of established cultured malignant ovarian epithelium on addition of ascites-derived vesicles was tested using a Matrigel-based invasion assay. Fractionation of malignant ascites revealed that extracellular matrix-degrading proteinases including MMPs and uPA are localized preferentially in membrane vesicles. All malignant vesicles tested, regardless of cancer stage, stimulated invasion. Furthermore, the combination of ovarian cancer cells and membrane vesicles resulted in greater uPA activation than that of cells or vesicles alone. Membrane vesicles from malignant ascites were also found to contain activated MMP-2, MMP-9, and uPA. Our data suggest that vesicle-stimulated proteinase activation leads to increased extracellular matrix degradation, which may facilitate tumor cell invasion and metastasis.
Collapse
Affiliation(s)
- Laura E Graves
- Department of Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
37
|
Prada M, Camporro J, Solís J, Fernández-Martínez R, Ferrer F. Laparotomía de second looken el tratamiento del áncer epitelial de ovario. CLINICA E INVESTIGACION EN GINECOLOGIA Y OBSTETRICIA 2004. [DOI: 10.1016/s0210-573x(04)77350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Davidson B, Risberg B, Reich R, Berner A. Effusion cytology in ovarian cancer: new molecular methods as aids to diagnosis and prognosis. Clin Lab Med 2003; 23:729-54, viii. [PMID: 14560537 DOI: 10.1016/s0272-2712(03)00058-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Carcinoma of the ovary is the leading cause of death from gynecologic cancer in western countries. Ovarian carcinoma is commonly associated with the accumulation of fluid that contains malignant cells in the peritoneal and pleural cavities. This article details the current knowledge regarding the diagnostic and biologic characteristics of ovarian carcinoma cells in effusions, and the genotypic and phenotypic differences between solid primary tumors and metastatic lesions. Finally, we present a new approach, by which the analysis of fresh frozen viable cells allows us to study in vivo the links between expression and activity of extracellular mediators, membrane receptors, intracellular signaling, and transcription factors, and their potential therapeutic and prognostic significance.
Collapse
Affiliation(s)
- Ben Davidson
- Department of Pathology, Norwegian Radium Hospital, University of Oslo, Montebello N-0310 Oslo, Norway.
| | | | | | | |
Collapse
|
39
|
von Gruenigen VE, Frasure HE, Reidy AM, Gil KM. Clinical disease course during the last year in ovarian cancer. Gynecol Oncol 2003; 90:619-24. [PMID: 13678735 DOI: 10.1016/s0090-8258(03)00418-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE(S) The objective was to determine whether there were changes in the pattern and nature of hospitalizations during the last year that could be used in the assessment of whether chemotherapy should be continued. METHODS Retrospective data were collected from patients who died from ovarian cancer between 1/2000 and 12/2001. Charts from four hospitals were reviewed to abstract chemotherapy, reason for hospitalization, and the incidence of three significant clinical events (bowel obstruction, pleural effusion requiring thoracentesis, and abdominal ascites requiring paracentesis). Data were analyzed in 3-month intervals. RESULTS Sixty-two patient charts were reviewed. Quarterly admissions increased linearly over the year (7, 18, 27, and 47, P < 0.0001). Hospitalizations for ascites, bowel obstruction, and pleural effusion began increasing around 6 months preceding death. Twenty-two patients did not receive chemotherapy during the last 3 months. Of the 40 patients receiving chemotherapy in the last 3 months, over half were not hospitalized during the period 4-6 months before death, and a further 20% were hospitalized for nonsignificant clinical events. Approximately one-quarter of the patients, however, continued to receive chemotherapy following hospitalization for a significant clinical event. CONCLUSION(S) There were significant changes in the pattern and nature of hospitalization during the last 6 months that included hospitalizations for bowel obstruction, pleural effusion, or ascites. The occurrence of these events suggests that further chemotherapy should be realistically evaluated with the patient, which may reduce the number of patients who receive chemotherapy during their last few months of life.
Collapse
Affiliation(s)
- Vivian E von Gruenigen
- University Hospitals of Cleveland/MacDonald Women's Hospital, Ireland Cancer Center, Case Western Reserve University, Cleveland, OH 44016, USA.
| | | | | | | |
Collapse
|
40
|
Chi DS, Liao JB, Leon LF, Venkatraman ES, Hensley ML, Bhaskaran D, Hoskins WJ. Identification of prognostic factors in advanced epithelial ovarian carcinoma. Gynecol Oncol 2001; 82:532-7. [PMID: 11520151 DOI: 10.1006/gyno.2001.6328] [Citation(s) in RCA: 181] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The Gynecologic Oncology Group (GOG) has demonstrated that age, tumor grade, and size and number of residual lesions after primary cytoreductive surgery are significant prognostic factors in advanced ovarian carcinoma. Recent studies have reported numerous other clinical features as having prognostic value. We sought to identify the independent prognostic factors for survival in a cohort of patients with advanced ovarian cancer. METHODS We performed a retrospective chart review of all patients with stage III and IV ovarian carcinoma who received their primary treatment at our institution between 1987 and 1994. RESULTS A total of 295 patients were identified, 282 of whom were evaluable. Of these 282 patients, 214 (76%) have died of disease or other causes. The median follow-up is 32 months (range: 1-139). Eighteen factors were evaluated for prognostic significance. Significant factors in univariate analysis included patient age, gravidity (0 vs > 0), parity (0 vs > 0), preoperative albumin level, preoperative total protein level, ascites (presence vs absence), disease stage (IIIA/IIIB vs IIIC vs IV), number of residual lesions (< or =20 vs >20), and diameter of largest residual tumor nodule (< or = 1 cm vs 1-2 cm vs > 2 cm). However, on multivariate analysis, only patient age (P < 0.001), ascites (P = 0.001), and size of residual disease (P = 0.005) retained prognostic significance. Substage of disease was of borderline significance (P = 0.086). CONCLUSION Although numerous clinical variables have recently been reported to have prognostic value in advanced ovarian carcinoma, only patient age, presence or absence of ascites, and diameter of the largest residual tumor nodule proved to be of statistical significance in our analysis.
Collapse
Affiliation(s)
- D S Chi
- Gynecology Service, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Prognosis of epithelial ovarian cancer related to its ascites. Chin J Cancer Res 2001. [DOI: 10.1007/s11670-001-0036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
42
|
Stallwood Y, Fisher KD, Gallimore PH, Mautner V. Neutralisation of adenovirus infectivity by ascitic fluid from ovarian cancer patients. Gene Ther 2000; 7:637-43. [PMID: 10800086 DOI: 10.1038/sj.gt.3301152] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animal models and phase I clinical trials have shown that repeat virus delivery and subsequent transgene expression is limited by the generation of humoral and cellular immune responses directed towards the therapeutic vector. The presence of a pre-existing immune response may even prevent initial delivery. In order to determine the presence of pre-existing anti-adenovirus humoral immunity we analysed ascitic fluid, collected from the peritoneal cavity of patients with advanced ovarian cancer. Twelve ascitic fluid and four matched serum samples were examined. The titre and isotype of anti-adenovirus antibodies was determined by ELISA, and Western blotting identified the molecular basis of the immune response, which was primarily directed towards fibre and penton base. Neutralisation of virus infectivity was assessed in vitro by measurement of green fluorescent protein reporter gene expression. We found that the ascitic fluid samples contain antibodies that recognise both adenovirus types 2 and 5, were predominantly IgG and directed towards the viral antigens responsible for cell adhesion, and had virus neutralising activity.
Collapse
Affiliation(s)
- Y Stallwood
- CRC Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TA, UK
| | | | | | | |
Collapse
|
43
|
Penson RT, Skates SJ, Fuller AJ, Seiden MV. Clinical course of stage IV epithelial ovarian cancer. J Clin Oncol 1999; 17:3361-2. [PMID: 10506641 DOI: 10.1200/jco.1999.17.10.3361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
44
|
Kapp KS, Kapp DS, Poschauko J, Stücklschweiger GF, Hackl A, Pickel H, Petru E, Winter R. The prognostic significance of peritoneal seeding and size of postsurgical residual in patients with stage III epithelial ovarian cancer treated with surgery, chemotherapy, and high-dose radiotherapy. Gynecol Oncol 1999; 74:400-7. [PMID: 10479500 DOI: 10.1006/gyno.1999.5477] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of this study was to retrospectively analyze the prognostic importance of age, histologic type and grade, ascites, lymph node status, size and type of postoperative residual disease, and radiation dose on disease-specific (DSS) and progression-free survival (PFS) in stage III epithelial ovarian cancer patients who had been treated with radical surgery, postoperative chemotherapy, and high-dose radiotherapy. METHODS Consolidation radiotherapy including whole abdominal radiation, pelvic, and upper abdominal boosts was employed in 46 patients who showed no evidence of residual or progressive disease after completion of multiagent chemotherapy. The median follow-up for all patients was 36 months and 103 months for patients at risk. The prognostic impact of pretreatment and treatment parameters on DSS and PFS was tested in univariate and multivariate analyses. RESULTS The 5-year DSS and PFS rates for all patients were 38 and 33%, and for patients with 0-< or =2 cm residual tumor 65 and 61%, respectively. In univariate analysis, initial peritoneal seeding (both: P = 0.02), ascites (P = 0.03; 0.01), size of residual (0-< or =2 cm vs >2 cm), and residual miliary subdiaphragmatic (MDS) and localized peritoneal seeding (LPS) in the upper abdomen (P = 0.0002; 0.0003) were significantly correlated with DSS and PFS. Dose of radiation (< or =30 vs >30 Gy) correlated with DSS only (P = 0.02). In multivariate analysis size of residual disease (0-< or =2 cm vs >2 cm and/or MDS or LPS) remained the only independent prognostic factor for DSS and PFS (both; P = 0. 001). CONCLUSION Patients with localized peritoneal seeding who were rendered free of disease elsewhere had an outcome equally poor as that of patients with gross residuals (>2 cm) in the upper abdomen. If our findings can be confirmed, attempted resection of all localized seeding in patients who are otherwise cytoreducible to no or minimal residual disease may be considered in combination with Taxol-containing regimens as are now being utilized for patients with gross disease.
Collapse
Affiliation(s)
- K S Kapp
- Department of Radiology, Karl-Franzens-University Medical School, Auenbruggerplatz 32, Graz, A-8036, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Schwartz PE, Rutherford TJ, Chambers JT, Kohorn EI, Thiel RP. Neoadjuvant chemotherapy for advanced ovarian cancer: long-term survival. Gynecol Oncol 1999; 72:93-9. [PMID: 9889037 DOI: 10.1006/gyno.1998.5236] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE The aim of this study was to compare the progression-free and overall survivals of women with advanced ovarian cancer treated with neoadjuvant chemotherapy followed by surgery with those treated conventionally with cytoreductive surgery followed by cytotoxic chemotherapy. MATERIALS AND METHODS Fifty-nine consecutive women with advanced malignancies compatible with ovarian cancer based on (1) physical examinations, (2) computerized tomography scans, and (3) cytologic or histologic specimens and treated with platinum-based combination chemotherapy, i.e., neoadjuvant chemotherapy, were retrospectively reviewed. Forty-one subsequently underwent cytoreductive surgery. Their overall and progression-free survivals were compared to those of 206 consecutive women with Stage IIIC and IV epithelial ovarian cancers treated with conventional cytoreductive surgery followed by platinum-based combination chemotherapy during the same era. RESULTS No statistical difference was observed in overall survival (P = 0.1578) or in progression-free survival between the group treated with neoadjuvant chemotherapy and the conventionally treated group (P = 0.5327) despite the neoadjuvant chemotherapy patients being statistically older (median age 67 years [range 44 to 85 years] vs a median age of 60 years [range 19 to 79 years] for conventionally treated patients; P < 0. 001) and having a statistically poorer performance status (P < 0. 001) than the conventionally treated group. Women undergoing cytoreductive surgery following neoadjuvant chemotherapy had a statistically improved overall survival (P < 0.0001) compared to those who did not undergo surgery. CONCLUSIONS Neoadjuvant chemotherapy does not compromise the survival of women treated for advanced ovarian cancer. Prospective randomized trials comparing neoadjuvant chemotherapy to conventional therapy to determine quality of life experiences and cost/benefit outcomes are now appropriate for women presenting with advanced ovarian cancer.
Collapse
Affiliation(s)
- P E Schwartz
- Department of Obstetrics and Gynecology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut, 06510, USA
| | | | | | | | | |
Collapse
|
46
|
De Simone GG. Treatment of Malignant Ascites. PROGRESS IN PALLIATIVE CARE 1999. [DOI: 10.1080/09699260.1999.11746825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
47
|
Beltran M, Fuentes R, Izquierdo A. Treatment of ovarian cancer with intraperitoneal cisplatin. N Engl J Med 1997; 336:1674; author reply 1675. [PMID: 9173271 DOI: 10.1056/nejm199706053362311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|