1
|
Wang Y, Zhu N, Liu J, Chen F, Song Y, Ma Y, Yang Z, Wang D. Role of tumor microenvironment in ovarian cancer metastasis and clinical advancements. J Transl Med 2025; 23:539. [PMID: 40369674 DOI: 10.1186/s12967-025-06508-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy worldwide, characterized by heterogeneity at the molecular, cellular and anatomical levels. Most patients are diagnosed at an advanced stage, characterized by widespread peritoneal metastasis. Despite optimal cytoreductive surgery and platinum-based chemotherapy, peritoneal spread and recurrence of OC are common, resulting in poor prognoses. The overall survival of patients with OC has not substantially improved over the past few decades, highlighting the urgent necessity of new treatment options. Unlike the classical lymphatic and hematogenous metastasis observed in other malignancies, OC primarily metastasizes through widespread peritoneal seeding. Tumor cells (the "seeds") exhibit specific affinities for certain organ microenvironments (the "soil"), and metastatic foci can only form when there is compatibility between the "seeds" and "soil." Recent studies have highlighted the tumor microenvironment (TME) as a critical factor influencing the interactions between the "seeds" and "soil," with ascites and the local peritoneal microenvironment playing pivotal roles in the initiation and progression of OC. Prior to metastasis, the interplay among tumor cells, immunosuppressive cells, and stromal cells leads to the formation of an immunosuppressive pre-metastatic niche in specific sites. This includes characteristic alterations in tumor cells, recruitment and functional anomalies of immune cells, and dysregulation of stromal cell distribution and function. TME-mediated crosstalk between cancer and stromal cells drives tumor progression, therapy resistance, and metastasis. In this review, we summarize the current knowledge on the onset and metastatic progression of OC. We provide a comprehensive discussion of the characteristics and functions of TME related to OC metastasis, as well as its association with peritoneal spread. We also outline ongoing relevant clinical trials, aiming to offer new insights for identifying potential effective biomarkers and therapeutic targets in future clinical practice.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Na Zhu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Jing Liu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China
| | - Fang Chen
- Department of Gynecology, People's Hospital of Liaoning Province, Shenyang, Liaoning Province, 110016, People's Republic of China
| | - Yang Song
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, People's Republic of China
| | - Yue Ma
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| | - Zhuo Yang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, People's Republic of China.
| |
Collapse
|
2
|
Deng S, Kim W, Cheng K, Yang Q, Singh Y, Bae G, Bézière N, Mager L, Kommoss S, Sprengel J, Trautwein C. Identification and impact of microbiota-derived metabolites in ascites of ovarian and gastrointestinal cancer. Cancer Metab 2025; 13:21. [PMID: 40361187 DOI: 10.1186/s40170-025-00391-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Malignant ascites is a common complication of advanced ovarian cancer (OC) and gastrointestinal cancer (GI), significantly impacting metastasis, quality of life, and survival. Increased intestinal permeability can lead to blood or lymphatic infiltration and microbial translocation from the gastrointestinal or uterine tract. This study aimed to identify microbiota-derived metabolites in ascites from OC (stages II-III and IV) and GI patients, assessing their roles in tumor progression. METHODS Malignant ascites samples from 18 OC and GI patients were analyzed using a four-dimensional (4D) untargeted metabolomics approach combining reversed-phase (RP) and hydrophilic interaction liquid chromatography (HILIC) with trapped ion mobility spectrometry time-of-flight mass spectrometry (timsTOF-MS). Additonally, a targeted flow cytometry-based cytokine panel was used to screen for inflammatory markers. Non-endogenous, microbiota-derived metabolites were identified through the Human Microbial Metabolome Database (MiMeDB). RESULTS OC stage IV exhibited metabolic profiles similar to GI cancers, while OC stage II-III differed significantly. Stage IV OC patients exhibited higher levels of 11 typically microbiome-derived metabolites, including 1-methylhistidine, 3-hydroxyanthranilic acid, 4-pyridoxic acid, biliverdin, butyryl-L-carnitine, hydroxypropionic acid, indole, lysophosphatidylinositol 18:1 (LPI 18:1), mevalonic acid, N-acetyl-L-phenylalanine, and nudifloramide, and lower levels of 5 metabolites, including benzyl alcohol, naringenin, o-cresol, octadecanedioic acid, and phenol, compared to stage II-III. Correlation analysis revealed positive associations between IL-10 and metabolites such as glucosamine and LPCs, while MCP-1 positively correlated with benzyl alcohol and phenol. CONCLUSION 4D metabolomics revealed distinct metabolic signatures in OC and GI ascites, highlighting microbiota-derived metabolites involved in lipid metabolism and inflammation. Metabolites like 3-hydroxyanthranilic acid, indole, and naringenin may serve as markers of disease progression and underscore the microbiota's role in shaping malignant ascites and tumor biology.
Collapse
Affiliation(s)
- Sisi Deng
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Wooyong Kim
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Kefan Cheng
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Qianlu Yang
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Gyuntae Bae
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Nicolas Bézière
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections", Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Lukas Mager
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections", Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Internal Medicine I, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Stefan Kommoss
- Department of Obstetrics and Gynecology, Diak Klinikum, Schäbisch Hall, Germany
| | - Jannik Sprengel
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Christoph Trautwein
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
- Core Facility Metabolomics, Faculty of Medicine, University of Tübingen, Tübingen, Germany.
- M3 Research Center for Microbiome, Metabolome and Malignome, Faculty of Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
3
|
Vierra MA, Morgan RB, Bhutiani N, White MG, Eng OS. Contemporary Management of Malignant Ascites. J Surg Res 2025; 307:157-175. [PMID: 40037156 DOI: 10.1016/j.jss.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/08/2024] [Accepted: 01/26/2025] [Indexed: 03/06/2025]
Abstract
INTRODUCTION Malignant ascites (MA) develops when malignant disease of the peritoneum causes excess fluid to accumulate in the abdominal cavity. It portends a poor prognosis and is associated with debilitating symptoms. While several palliative therapies exist, none have proven curative or free from side effects and complications. This review article describes experimental therapies on the horizon and the contemporary management of MA. MATERIALS AND METHODS A literature review was performed using MEDLINE/PubMed, in which studies of emerging or experimental therapies under investigation for the management of MA were reviewed. Current therapies were also reviewed to provide important context. Data, including study design, sample size, primary and secondary outcomes, and side effects were recorded and described. Studies were then categorized into distinct sections and subsections, with tables corresponding to each section. RESULTS Five current therapies, including paracentesis, diuretics, peritoneovenous shunting, permanent catheters, and intraperitoneal chemotherapy, are described. Their limitations in effectively managing MA are highlighted. The "Experimental therapies" section is subsectioned into several categories, with the major studies corresponding to each section thoroughly described regarding methods, results, and validity. A final section describes treatments for mucinous ascites, which has distinct characteristics. CONCLUSIONS While each of the experimental therapies described offers unique benefits and has demonstrated some promise in managing MA, they all have limitations that have thus far prevented any one of them from being routinely used in practice. MA remains a challenging condition to treat, warranting further research into novel therapies.
Collapse
Affiliation(s)
- Mason A Vierra
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Ryan B Morgan
- Department of Surgery, University of Chicago Medical Center, Chicago, Illinois
| | - Neal Bhutiani
- Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Michael G White
- Department of Colon & Rectal Surgery, MD Anderson Cancer Center, Houston, Texas
| | - Oliver S Eng
- Department of Surgery, University of California Irvine, Orange, California
| |
Collapse
|
4
|
Berger JM, Lötsch F, Berghoff AS, Lamm WW, Preusser M, Jeryczynski G. A case of fungal peritonitis in a patient with paramalignant ascites. Med Mycol Case Rep 2024; 45:100660. [PMID: 39149598 PMCID: PMC11325765 DOI: 10.1016/j.mmcr.2024.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
Here, we present the case of a patient with a metastatic neuroendocrine tumor with cytologically negative ascites treated for spontaneous bacterial peritonitis (SBP). Ascitic cultures remained negative for bacterial growth but were positive for Candida albicans 8 days after SBP diagnosis. ß-D-glucan was only positive in ascites, while being negative in blood. Blood cultures remained negative throughout the whole admission. Fungal peritonitis presumably originated from an impending bowl perforation or an increasing vascular permeability caused by an increase in VEGF secondary to diffuse infiltration by the underlying malignant disease.
Collapse
Affiliation(s)
- Julia M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Felix Lötsch
- Clinical Division of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Wolfgang W Lamm
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Georg Jeryczynski
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Pampeno C, Opp S, Hurtado A, Meruelo D. Sindbis Virus Vaccine Platform: A Promising Oncolytic Virus-Mediated Approach for Ovarian Cancer Treatment. Int J Mol Sci 2024; 25:2925. [PMID: 38474178 PMCID: PMC10932354 DOI: 10.3390/ijms25052925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This review article provides a comprehensive overview of a novel Sindbis virus vaccine platform as potential immunotherapy for ovarian cancer patients. Ovarian cancer is the most lethal of all gynecological malignancies. The majority of high-grade serous ovarian cancer (HGSOC) patients are diagnosed with advanced disease. Current treatment options are very aggressive and limited, resulting in tumor recurrences and 50-60% patient mortality within 5 years. The unique properties of armed oncolytic Sindbis virus vectors (SV) in vivo have garnered significant interest in recent years to potently target and treat ovarian cancer. We discuss the molecular biology of Sindbis virus, its mechanisms of action against ovarian cancer cells, preclinical in vivo studies, and future perspectives. The potential of Sindbis virus-based therapies for ovarian cancer treatment holds great promise and warrants further investigation. Investigations using other oncolytic viruses in preclinical studies and clinical trials are also presented.
Collapse
Affiliation(s)
- Christine Pampeno
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| | | | - Alicia Hurtado
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Daniel Meruelo
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| |
Collapse
|
6
|
Zhang J, Qi Z, Ou W, Mi X, Fang Y, Zhang W, Yang Z, Zhou Y, Lin X, Hou J, Yuan Z. Advances in the treatment of malignant ascites in China. Support Care Cancer 2024; 32:97. [PMID: 38200158 DOI: 10.1007/s00520-023-08299-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
PURPOSE Malignant ascites (MA) often occurs in recurrent abdominal malignant tumors, and the large amount of ascites associated with cancerous peritonitis not only leads to severe abdominal distension and breathing difficulties, but also reduces the patient's quality of life and ability to resist diseases, which usually makes it difficult to carry out anti-cancer treatment. The exploration of MA treatment methods is also a key link in MA treatment. This article is going to review the treatment of MA, to provide details for further research on the treatment of MA, and to provide some guidance for the clinical treatment of MA. METHOD This review analyzes various expert papers and summarizes them to obtain the paper. RESULT There are various treatment methods for MA, including systemic therapy and local therapy. Among them, systemic therapy includes diuretic therapy, chemotherapy, immunotherapy, targeted therapy, anti angiogenic therapy, CAR-T, and vaccine. Local therapy includes puncture surgery, peritoneal vein shunt surgery, acellular ascites infusion therapy, radioactive nuclide intraperitoneal injection therapy, tunnel catheter, and intraperitoneal hyperthermia chemotherapy. And traditional Chinese medicine treatment has also played a role in enhancing efficacy and reducing toxicity to a certain extent. CONCLUSION Although there has been significant progress in the treatment of MA, it is still one of the clinical difficulties. Exploring the combination or method of drugs with the best therapeutic effect and the least adverse reactions to control MA is still an urgent problem to be solved.
Collapse
Affiliation(s)
- Junzi Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhaoxue Qi
- Department of Secretory Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Wenjie Ou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuguang Mi
- Department of Central Laboratory, Jilin Provincial People's Hospital, Changchun, China
| | - Yanqiu Fang
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, China
| | - Wenqi Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhen Yang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ying Zhou
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, China
| | - Xiuying Lin
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, China
| | - Junjie Hou
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, China.
| | - Zhixin Yuan
- Department of Emergency Surgery, Jilin Provincial People's Hospital, Changchun, China.
| |
Collapse
|
7
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
8
|
Berger JM, Alany A, Berchtold L, Puhr R, Friedrich A, Scheiner B, Prager GW, Preusser M, Berghoff AS, Bergen ES. Prognosticators of survival in patients with metastatic pancreatic cancer and ascites. ESMO Open 2023; 8:102048. [PMID: 37977000 PMCID: PMC10774951 DOI: 10.1016/j.esmoop.2023.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Identification of factors associated with survival after ascites diagnosis in metastatic pancreatic cancer (mPC) patients may guide treatment decisions and help to maintain quality of life in this highly symptomatic patient collective. PATIENTS AND METHODS All patients treated for mPC at the Medical University of Vienna between 2010 and 2019 developing ascites throughout their course of disease were identified by retrospective chart review. General risk factors, metastatic sites, systemic inflammation and liver function parameters, as well as type of treatment after ascites diagnosis were investigated for associations with survival. RESULTS One hundred and seventeen mPC patients with ascites were included in this study. Median time from mPC to ascites diagnosis was 8.9 months (range 0-99 months) and median overall survival (OS) after ascites diagnosis was 27.4 days (range 21.3-42.6 days). Identified prognostic factors at ascites diagnosis independently associated with an impaired OS were presence of liver metastases [hazard ratio (HR): 2.07, 95% confidence interval (CI) 1.13-3.79, P = 0.018), peritoneal carcinomatosis (HR: 1.74, 95% CI 1.11-2.71, P = 0.015), and portal vein obstruction (HR: 2.52, 95% CI 1.29-4.90, P = 0.007). Compared with best supportive care, continuation of systemic therapy after ascites diagnosis was independently associated with survival (HR: 0.35, 95% CI 0.20-0.61, P < 0.001) with a median OS of 62 days (95% CI 51-129 days, P < 0.001) versus 16 days (95% CI 11-24 days), respectively. CONCLUSIONS Liver and peritoneal metastases as well as portal vein obstruction were found to be prognostic factors after ascites diagnosis in mPC patients. Continuation of systemic therapy after ascites diagnosis was associated with a longer OS, which needs to be evaluated in larger clinical trials including quality-of-life assessment.
Collapse
Affiliation(s)
- J M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Alany
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - L Berchtold
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - R Puhr
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Friedrich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - B Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - G W Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - M Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - E S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna.
| |
Collapse
|
9
|
Berger JM, Preusser M, Berghoff AS, Bergen ES. Malignant ascites: Current therapy options and treatment prospects. Cancer Treat Rev 2023; 121:102646. [PMID: 39492370 DOI: 10.1016/j.ctrv.2023.102646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/05/2024]
Abstract
Ascites formation is a common complication of cancer with a significant symptomatic burden for patients. Malignant ascites (MA) is defined by the presence of tumor cells within the ascitic fluid. It does not only cause substantial morbidity, but is also associated with impaired survival. Considering the frequent occurrence of MA, it still represents a clinical challenge for physicians with limited therapy options, mainly comprising of the treatment of the primary tumor and effusion drainage. Particularly the lack of pathophysiological insight limits the development of effective, causative therapies. Causes of MA development such as lymphatic vessel obstruction and the effects of tumor secreted vascular endothelial growth factor (VEGF) have been known for decades. Novel research suggests that the intraperitoneal immune system may also induce and maintain MA accumulation. In this review, we assess current knowledge on the pathophysiology of MA and summarize available evidence of treatment approaches. Also, factors contributing to ascites formation without proof of tumor cells in the peritoneal cavity, defined as paramalignant ascites, with potential treatment strategies are discussed. We further focus on novel findings in the pathophysiology of MA that might lead to treatment improvement in the near future and discussed relevant knowledge gaps in this field.
Collapse
Affiliation(s)
- Julia M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elisabeth S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
10
|
Berger J, Alany A, Puhr R, Berchtold L, Friedrich A, Scheiner B, Prager G, Berghoff A, Preusser M, Bergen E. Clinical risk factors for ascites in metastatic pancreatic cancer. ESMO Open 2023; 8:101200. [PMID: 36989885 PMCID: PMC10163163 DOI: 10.1016/j.esmoop.2023.101200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Malignant ascites is common in metastatic pancreatic cancer (mPC) and its management still remains a clinical challenge. Early identification of patients at risk for ascites development may support and guide treatment decisions. MATERIALS AND METHODS Data of patients treated for mPC at the Medical University of Vienna between 2010 and 2019 were collected by retrospective chart review. Ascites was defined as clinically relevant accumulation of intraperitoneal fluid diagnosed by ultrasound or computer tomography scan of the abdomen. We investigated the association between general risk factors, metastatic sites, liver function, systemic inflammation as well as portal vein obstruction (PVO) and ascites development. RESULTS Among 581 patients with mPC included in this study, 122 (21.0%) developed ascites after a median of 8.7 months after diagnosis of metastatic disease. The occurrence of ascites led to an 8.9-fold increased risk of death [confidence interval (CI) 7.2-11, P < 0.001] with a median overall survival of 1 month thereafter. Clinical risk factors for ascites were male sex [hazard ratio (HR) 1.71, CI 1.00-2.90, P = 0.048], peritoneal carcinomatosis (HR 6.79, CI 4.09-11.3, P < 0.001), liver metastases (HR 2.16, CI 1.19-3.91, P = 0.011), an albumin-bilirubin (ALBI) score grade 3 (HR 6.79, CI 2.11-21.8, P = 0.001), PVO (HR 2.28, CI 1.15-4.52, P = 0.019), and an elevated C-reactive protein (CRP) (HR 4.19, CI 1.58-11.1, P = 0.004). CONCLUSIONS Survival after diagnosis of ascites is very limited in mPC patients. Male sex, liver and peritoneal metastases, impaired liver function, PVO, as well as systemic inflammation were identified as independent risk factors for ascites development in this uniquely large real-life patient cohort.
Collapse
|
11
|
Han MY, Borazanci EH. Malignant ascites in pancreatic cancer: Pathophysiology, diagnosis, molecular characterization, and therapeutic strategies. Front Oncol 2023; 13:1138759. [PMID: 37007072 PMCID: PMC10060830 DOI: 10.3389/fonc.2023.1138759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Malignant ascites is the accumulation of fluid in the peritoneum as a result of advanced cancer and often signifies the terminal phase of the disease. Management of malignant ascites remains a clinical challenge as symptom palliation is the current standard of cure. Previously, studies examining malignant ascites largely focused on ovarian and gastric cancer. In recent years, there has been a significant increase in research on malignant ascites in pancreatic cancer. Malignant ascites is usually diagnosed based on positive cytology, but cytology is not always diagnostic, indicating the need for novel diagnostic tools and biomarkers. This review aims to summarize the current understanding of malignant ascites in pancreatic cancer and the recent advances in the molecular characterization of malignant ascites fluid from patients with pancreatic cancer including analysis of soluble molecules and extracellular vesicles. Current standard of care treatment options such as paracenteses and diuretics are outlined along with new emerging treatment strategies such as immunotherapy and small-molecule based therapies. New potential investigative directions resulting from these studies are also highlighted.
Collapse
Affiliation(s)
- Margaret Y. Han
- Department of Biosciences, Rice University, Houston, TX, United States
| | - Erkut H. Borazanci
- Department of Oncology, HonorHealth Research Institute, Scottsdale, AZ, United States
- *Correspondence: Erkut H. Borazanci,
| |
Collapse
|
12
|
Sharma T, Nisar S, Masoodi T, Macha MA, Uddin S, Akil AAS, Pandita TK, Singh M, Bhat AA. Current and emerging biomarkers in ovarian cancer diagnosis; CA125 and beyond. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:85-114. [PMID: 36707207 DOI: 10.1016/bs.apcsb.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Ovarian cancer (OC) is one of the most common causes of cancer-related death in women worldwide. Its five-year survival rates are worse than the two most common gynecological cancers, cervical and endometrial. This is because it is asymptomatic in the early stages and usually detected in the advanced metastasized stage. Thus, survival is increasingly dependent on timely diagnosis. The delay in detection is contributed partly by the occurrence of non-specific clinical symptoms in the early stages and the lack of effective biomarkers and detection approaches. This underlines the need for biomarker identification and clinical validation, enabling earlier diagnosis, effective prognosis, and response to therapy. Apart from the traditional diagnostic biomarkers for OC, several new biomarkers have been delineated using advanced high-throughput molecular approaches in recent years. They are currently being clinically evaluated for their true diagnostic potential. In this chapter, we document the commonly utilized traditional screening markers and recently identified emerging biomarkers in OC diagnosis, focusing on secretory and protein biomarkers. We also briefly reviewed the recent advances and prospects in OC diagnosis.
Collapse
Affiliation(s)
- Tarang Sharma
- Department of Medical Oncology, Dr. B.R Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sabah Nisar
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Cancer immunology and genetics, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ammira Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, United States
| | - Mayank Singh
- Department of Medical Oncology, Dr. B.R Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
13
|
Al-Marzouki L, Stavrakos VS, Pal S, Giannias B, Bourdeau F, Rayes R, Bertos N, Najmeh S, Spicer JD, Cools-Lartigue J, Bailey SD, Ferri L, Sangwan V. Soluble factors in malignant ascites promote the metastatic adhesion of gastric adenocarcinoma cells. Gastric Cancer 2023; 26:55-68. [PMID: 36059037 DOI: 10.1007/s10120-022-01338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/25/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Adenocarcinoma of the proximal stomach is the fastest rising malignancy in North America. It is commonly associated with peritoneal accumulation of malignant ascites (MA), a fluid containing cancer and inflammatory cells and soluble proteins. Peritoneal metastasis (PM) is the most common site of gastric cancer (GC) progression after curative-intent surgery and is the leading cause of death among GC patients. METHODS/RESULTS Using a panel of gastric adenocarcinoma cell lines (human: MKN 45, SNU-5; murine: NCC-S1M), we demonstrate that prior incubation of GC cells with MA results in a significant (> 1.7-fold) increase in the number of cells capable of adhering to human peritoneal mesothelial cells (HPMC) (p < 0.05). We then corroborate these findings using an ex vivo PM model and show that MA also significantly enhances the ability of GC cells to adhere to strips of human peritoneum (p < 0.05). Using a multiplex ELISA, we identify MIF and VEGF as consistently elevated across MA samples from GC patients (p < 0.05). We demonstrate that agents that block the effects of MIF or VEGF abrogate the ability of MA to stimulate the adhesion of GC cells to adhere to human peritoneum and promote both ex vivo and in vivo metastases. CONCLUSION Agents targeting MIF or VEGF may be relevant to the treatment or prevention of PM in GC patients.
Collapse
Affiliation(s)
- Luai Al-Marzouki
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vivian S Stavrakos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Sanjima Pal
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Betty Giannias
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - France Bourdeau
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Roni Rayes
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Sara Najmeh
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan D Spicer
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan Cools-Lartigue
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Swneke D Bailey
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Lorenzo Ferri
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada.
| | - Veena Sangwan
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Sterling C, Márquez-Garbán D, Vadgama JV, Pietras RJ. Squalamines in Blockade of Tumor-Associated Angiogenesis and Cancer Progression. Cancers (Basel) 2022; 14:5154. [PMID: 36291938 PMCID: PMC9601113 DOI: 10.3390/cancers14205154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/30/2022] Open
Abstract
Mechanisms of action of squalamine in human vascular endothelial cells indicate that this compound attaches to cell membranes, potentially interacting with calmodulin, Na+/H+ exchanger isoform NHE3 and other signaling pathways involved in the angiogenic process. Thus, squalamine elicits blockade of VEGF-induced endothelial tube-like formation in vitro. Further, squalamine reduces growth of several preclinical models of human cancers in vivo and acts to stop metastatic tumor spread, actions due largely to blockade of angiogenesis induced by the tumor and tumor microenvironment. Squalamine in Phase I/II trials, alone or combined with standard care, shows promising antitumor activity with limited side-effects in patients with advanced solid cancers. Increased attention on squalamine regulation of signaling pathways with or without combination treatments in solid malignancies deserves further study.
Collapse
Affiliation(s)
- Colin Sterling
- Division of Cancer Research and Training, Charles Drew University School of Medicine and Science, Los Angeles, CA 90059, USA
| | - Diana Márquez-Garbán
- Division of Hematology-Oncology, Department of Medicine, UCLA David Geffen School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Charles Drew University School of Medicine and Science, Los Angeles, CA 90059, USA
- Division of Hematology-Oncology, Department of Medicine, UCLA David Geffen School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Richard J. Pietras
- Division of Cancer Research and Training, Charles Drew University School of Medicine and Science, Los Angeles, CA 90059, USA
- Division of Hematology-Oncology, Department of Medicine, UCLA David Geffen School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Salati M, Caputo F, Bocconi A, Cerri S, Baldessari C, Piacentini F, Dominici M, Gelsomino F. Successes and failures of angiogenesis blockade in gastric and gastro-esophageal junction adenocarcinoma. Front Oncol 2022; 12:993573. [PMID: 36212393 PMCID: PMC9540203 DOI: 10.3389/fonc.2022.993573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric and gastro-esophageal junction adenocarcinoma (GEA) remains a considerable major public health problem worldwide, being the fifth most common cancer with a fatality-to-case ratio that stands still at 70%. Angiogenesis, which is a well-established cancer hallmark, exerts a fundamental role in cancer initiation and progression and its targeting has been actively pursued as a promising therapeutic strategy in GEA. A wealth of clinical trials has been conducted, investigating anti-angiogenic agents including VEGF-directed monoclonal antibodies, small molecules tyrosine kinase inhibitors and VEGF-Trap agents both in the resectable and advanced setting, reporting controversial results. While phase III randomized trials testing the anti-VEGFR-2 antibody Ramucirumab and the selective VEGFR-2 tyrosine kinase inhibitor Apatinib demonstrated a significant survival benefit in later lines, the shift of angiogenesis inhibitors in the perioperative and first-line setting failed to improve patients' outcome in GEAs. The molecular landscape of disease, together with novel combinatorial strategies and biomarker-selected approaches are under investigation as key elements to the success of angiogenesis blockade in GEA. In this article, we critically review the existing literature on the biological rationale and clinical development of antiangiogenic agents in GEA, discussing major achievements, limitations and future developments, aiming at fully realizing the potential of this therapeutic approach.
Collapse
Affiliation(s)
- Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
- PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Caputo
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Alessandro Bocconi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Sara Cerri
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Federico Piacentini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| |
Collapse
|
16
|
Transcending Blood—Opportunities for Alternate Liquid Biopsies in Oncology. Cancers (Basel) 2022; 14:cancers14051309. [PMID: 35267615 PMCID: PMC8909855 DOI: 10.3390/cancers14051309] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cell-free DNA—DNA that has been expelled from cells and can be isolated from blood plasma and other body fluids—is a useful tool in medicine, with applications as a biomarker in diagnosis, prognosis, disease profiling, and treatment selection. In oncology, the ease of access to the tumour genome is a major advantage of cell-free DNA, but while this has led to significant research in blood, other body fluids have not received equal attention. This review article summarises the current research into cell-free DNA in non-blood body fluids, highlighting its values and limitations, and suggesting the direction of future studies. We conclude that cell-free DNA from non-blood body fluids may provide additional information to supplement traditional biopsies, allowing informative and improved patient care across many cancer types. Abstract Cell-free DNA (cfDNA) is a useful molecular biomarker in oncology research and treatment, but while research into its properties in blood has flourished, there remains much to be discovered about cfDNA in other body fluids. The cfDNA from saliva, sputum, cerebrospinal fluid, urine, faeces, pleural effusions, and ascites has unique advantages over blood, and has potential as an alternative ‘liquid biopsy’ template. This review summarises the state of current knowledge and identifies the gaps in our understanding of non-blood liquid biopsies; where their advantages lie, where caution is needed, where they might fit clinically, and where research should focus in order to accelerate clinical implementation. An emphasis is placed on ascites and pleural effusions, being pathological fluids directly associated with cancer. We conclude that non-blood fluids are viable sources of cfDNA in situations where solid tissue biopsies are inaccessible, or only accessible from dated archived specimens. In addition, we show that due to the abundance of cfDNA in non-blood fluids, they can outperform blood in many circumstances. We demonstrate multiple instances in which DNA from various sources can provide additional information, and thus we advocate for analysing non-blood sources as a complement to blood and/or tissue. Further research into these fluids will highlight opportunities to improve patient outcomes across cancer types.
Collapse
|
17
|
Conrad C, Moore K, Polacheck W, Rizvi I, Scarcelli G. Mechanical Modulation of Ovarian Cancer Tumor Nodules Under Flow. IEEE Trans Biomed Eng 2022; 69:294-301. [PMID: 34170820 PMCID: PMC8750319 DOI: 10.1109/tbme.2021.3092641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Perfusion models are valuable tools to mimic complex features of the tumor microenvironment and to study cell behavior. In ovarian cancer, mimicking disease pathology of ascites has been achieved by seeding tumor nodules on a basement membrane and subjecting them to long-term continuous flow. In this scenario it is particularly important to study the role of mechanical stress on cancer progression. Mechanical cues are already known to be important in key cancer processes such as survival, proliferation, and migration. However, probing cell mechanical properties within microfluidic platforms has not been achievable with current technologies since samples are not easily accessible within most microfluidic channels. METHODS Here, to analyze the mechanical properties of cells within a perfusion chamber, we use Brillouin confocal microscopy, an all-optical technique that requires no contact or perturbation to the sample. RESULTS Our results indicate that ovarian cancer nodules under long-term continuous flow have a significantly lower longitudinal modulus compared to nodules maintained in a static condition. CONCLUSION We further dissect the role of distinct mechanical perturbations (e.g., shear flow, osmolality) on tumor nodule properties. SIGNIFICANCE In summary, the unique combination of a long-term microfluidic culture and noninvasive mechanical analysis technique provides insights on the effects of physical forces in ovarian cancer pathology.
Collapse
|
18
|
Kamposioras K, Geraghty J, Appleyard J, Dawod M, Papadimitriou K, Lamarca A, Anthoney A. Pancreaticobiliary Malignancies in the Emergency Room: Management of Acute Complications and Oncological Emergencies. J Gastrointest Cancer 2021; 53:1050-1065. [PMID: 34648136 PMCID: PMC9630225 DOI: 10.1007/s12029-021-00718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 11/29/2022]
Abstract
Background Management of pancreaticobiliary (PB) malignancies remains a clinical challenge. In this review, we focus on the management of oncological emergencies in PB malignancies and the potential complication of associated therapeutic interventions. Methods Biobliographic review of current evidence on the management of oncological emergencies, their potential complications, as well as synthesis of recommendations was performed. The pathogenesis, frequency, related symptoms as well as appropriate investigations are presented. Results The oncologic emergencies in PB patients were summarised in six categories: (1) hematological (including febrile neutropaenia, thrombocytopenia, coagulopathies), (2) gastrointestinal (gastric outlet and biliary obstruction, gastrointestinal bleeding), (3) thromboembolic events, (4) ascites, (5) metabolic disorders and (6) neurologic complications. The pathogenesis, frequency, related symptoms as well as appropriate investigations are also presented. Conclusion Patients with PB malignancies are at increased risk of a wide variation of medical emergencies. Clinical knowledge, early recognition and collaboration with the relevant specialties are critical to manage these complications effectively, tailoring overall management around the actual prognosis and individuals’ expectations.
Collapse
Affiliation(s)
| | - Joe Geraghty
- Department of Gastroenterology, Manchester Royal Infirmary, Manchester, UK
| | | | - Mohammed Dawod
- The Christie NHS Foundation Trust, Manchester, Greater Manchester, UK
| | | | - Angela Lamarca
- The Christie NHS Foundation Trust, Manchester, Greater Manchester, UK.,Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alan Anthoney
- Leeds Institute of Medical Research, St James' Institute of Oncology, St James' University Hospital, University of Leeds, Leeds, LS9 7TF, UK.
| |
Collapse
|
19
|
Pitman M, Oehler MK, Pitson SM. Sphingolipids as multifaceted mediators in ovarian cancer. Cell Signal 2021; 81:109949. [PMID: 33571664 DOI: 10.1016/j.cellsig.2021.109949] [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] [Received: 10/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is the most lethal gynaecological malignancy. It is commonly diagnosed at advanced stage when it has metastasised to the abdominal cavity and treatment becomes very challenging. While current standard therapy involving debulking surgery and platinum + taxane-based chemotherapy is associated with high response rates initially, the large majority of patients relapse and ultimately succumb to chemotherapy-resistant disease. In order to improve survival novel strategies for early detection and therapeutics against treatment-refractory disease are urgently needed. A promising new target against ovarian cancer is the sphingolipid pathway which is commonly hijacked in cancer to support cell proliferation and survival and has been shown to promote chemoresistance and metastasis in a wide range of malignant neoplasms. In particular, the sphingosine kinase 1-sphingosine 1-phosphate receptor 1 axis has been shown to be altered in ovarian cancer in multiple ways and therefore represents an attractive therapeutic target. Here we review the roles of sphingolipids in ovarian cancer progression, metastasis and chemoresistance, highlighting novel strategies to target this pathway that represent potential avenues to improve patient survival.
Collapse
Affiliation(s)
- MelissaR Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5000, Australia.
| | - Martin K Oehler
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
20
|
Quan Q, Zhou S, Liu Y, Yin W, Liao Q, Ren S, Zhang F, Meng Y, Mu X. Relationship between ascites volume and clinical outcomes in epithelial ovarian cancer. J Obstet Gynaecol Res 2021; 47:1527-1535. [PMID: 33506580 DOI: 10.1111/jog.14682] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Ascites is a tumor microenvironment, ascites and massive ascites-induce compression could promote the progression of epithelial ovarian cancer (EOC); however, the impact of ascites volume on clinical outcomes has not been studied extensively. We aimed to investigate the association between ascites volume and clinical outcomes especially platinum resistance in EOC. METHODS We retrospectively evaluated a total of 546 EOC patients with respect to the amount of ascites, clinicopathologic factors, and survival. Using the threshold of 1500 ml to classify patients into small- and large-volume ascites groups, we analyzed the correlation between ascites volume and clinicopathological factors, including platinum-free interval (PFI), and prognosis. RESULTS Patients with large volume ascites were more likely to present with later stage disease, primary platinum-resistant (PPR) cancer, and suboptimal cytoreduction. Prolonged PFI was associated with decreased ascites volume. The large-volume ascites group showed worse progression-free survival (PFS) and overall survival (OS). An increase in ascites volume was associated with an increased risk of disease recurrence (hazard ratio [HR] = 1.115, 95% confidence interval [CI]: 1.035-1.200) and death (HR = 1.213, 95% CI: 1.090-1.350). CONCLUSIONS Ascites was an independent predictor of PFS and OS in EOC patients. A large volume of ascites predicated a shortened PFI, an increased incidence of PPR and suboptimal cytoreduction. Thus, the volume of ascites is a simply available clinical parameter, which could be used to evaluate the prognosis and platinum resistance of EOC patients early, it contributes to formulate individualized treatment plan and improve the outcome of EOC patients.
Collapse
Affiliation(s)
- Quan Quan
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuwei Zhou
- Department of Obstetrics, Chongqing Health Center for Women and Children, Chongqing, China
| | - Yao Liu
- Department of Obstetrics and Gynecology, Chengdu First People Hospital, Chengdu, Sichuan, China
| | - Wanchun Yin
- Department of Gynecology, The First People's Hospital of Chongqing Liangjiang New Area, Chongqing, China
| | - Qianqian Liao
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Siling Ren
- Department of Obstetrics, Chongqing Fuling District Maternal and Child Health Care, Chongqing, China
| | - Fenfen Zhang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Meng
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoling Mu
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Liu M, Silva-Sanchez A, Randall TD, Meza-Perez S. Specialized immune responses in the peritoneal cavity and omentum. J Leukoc Biol 2020; 109:717-729. [PMID: 32881077 DOI: 10.1002/jlb.5mir0720-271rr] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/13/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022] Open
Abstract
The peritoneal cavity is a fluid filled space that holds most of the abdominal organs, including the omentum, a visceral adipose tissue that contains milky spots or clusters of leukocytes that are organized similar to those in conventional lymphoid tissues. A unique assortment of leukocytes patrol the peritoneal cavity and migrate in and out of the milky spots, where they encounter Ags or pathogens from the peritoneal fluid and respond accordingly. The principal role of leukocytes in the peritoneal cavity is to preserve tissue homeostasis and secure tissue repair. However, when peritoneal homeostasis is disturbed by inflammation, infection, obesity, or tumor metastasis, specialized fibroblastic stromal cells and mesothelial cells in the omentum regulate the recruitment of peritoneal leukocytes and steer their activation in unique ways. In this review, the types of cells that reside in the peritoneal cavity, the role of the omentum in their maintenance and activation, and how these processes function in response to pathogens and malignancy will be discussed.
Collapse
Affiliation(s)
- Mingyong Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
22
|
Sabbion RO, Terra RM, Teixeira LR, Acencio MMP, Augusto MC, Costa PB, Fernandes PMP. Influence of the progression of pleural neoplasia on the outcome of pleurodesis in mice. Oncotarget 2020; 11:2002-2009. [PMID: 32523654 PMCID: PMC7260117 DOI: 10.18632/oncotarget.27610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/19/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose: Experimental study aimed at evaluating whether pleural neoplastic disease is associated with the degree of pleural fibrosis over time caused by talc pleurodesis. The study describes changes in levels of inflammatory mediators and determines whether the course of time involved in progression of neoplastic pleural disease is the factor that influences safety of talc pleurodesis usage in mice. Materials and Methods: Animals were randomized into two groups: Cancer group (CG) that received intrapleural injection of Lewis cells or Saline group (SG) that received saline injection. After, the animals were subdivided into Early (pleurodesis 3 days after pleural injection) and Late (pleurodesis 7 days after pleural injection) groups. Half of the animals in each group were euthanized 24 hours after pleurodesis (to obtain the inflammatory data); the remaining animals were killed after 8 days (to obtain the scores of pleural fibrosis). Results: CGs had lower fibrosis scores than SGs comparing early phases to late phases. Inflammation scores were lower in CGs, particularly in Late group. In SGs the inflammation was intense in 100% of the animals. In Late CG group pleural adhesions had the lowest scores; we found intense fibrosis only in SGs. VEGF and LDH levels had increased in animals with cancer, particularly in Late group. Systemic distribution of talc occurred only in Late CG. Conclusions: The time for pleural neoplasia to evolve is inversely proportional to the degree of pleural fibrosis. Earlier pleurodesis yielded the best results related to fibrosis, with less systemic inflammation and is safer in mice.
Collapse
Affiliation(s)
- Rodrigo Olivio Sabbion
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ricardo Mingarini Terra
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Lisete Ribeiro Teixeira
- Laboratorio de Pleura-Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Milena Marques Pagliarelli Acencio
- Laboratorio de Pleura-Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Marcia Cristina Augusto
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Priscila Berenice Costa
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Paulo Manuel Pego Fernandes
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
23
|
Sha R, Badhulika S. Recent advancements in fabrication of nanomaterial based biosensors for diagnosis of ovarian cancer: a comprehensive review. Mikrochim Acta 2020; 187:181. [PMID: 32076837 DOI: 10.1007/s00604-020-4152-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/02/2020] [Indexed: 12/30/2022]
Abstract
Ovarian cancer is commonly diagnosed via determination of biomarkers like CA125, Mucin 1, HE4, and prostasin that can be present in the blood. However, there is a substantial need for less expensive, simpler, and portable diagnostic tools, both for timely diagnosis and management of ovarian cancer. This review (with 101 refs.) discusses various kinds of nanomaterial-based biosensors for tumor markers. Following an introduction into the field, a first section covers different kinds of biomarkers for ovarian cancer including CA125 (MUC16), mucin 1 (MUC1), human epididymis protein 4 (HE4), and prostasin. This is followed by a short overview on conventional diagnostic approaches. A large section is then presented on biosensors for determination of ovarian cancer, with subsections on optical biosensors (fluorimetric, colorimetric, surface plasmon resonance, chemiluminescence, electrochemiluminescence), on electrochemical sensors, molecularly imprinted sensors, paper-based biosensors, microfluidic (lab-on-a-chip) assays, chemiresistive and field effect transistor-based sensors, and giant magnetoresistive sensors. Tables are presented that give an overview on the wealth of methods and materials. A concluding section summarizes the current status, addresses current challenges, and gives an outlook on potential future trends. Graphical abstract Schematic representation of the review covering the advancements in the fabrication of various nanomaterial based biosensors for diagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Rinky Sha
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad, 502285, India
| | - Sushmee Badhulika
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad, 502285, India.
| |
Collapse
|
24
|
Yamasaki F, Kolakshyapati M, Takano M, Yonezawa U, Nishibuchi I, Imano N, Taguchi A, Onishi S, Amatya VJ, Takeshima Y, Nagata Y, Kurisu K, Sugiyama K. Effect of bevacizumab against cystic components of brain tumors. Cancer Med 2019; 8:6519-6527. [PMID: 31498567 PMCID: PMC6825995 DOI: 10.1002/cam4.2537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022] Open
Abstract
Background Bevacizumab improves symptoms via reducing the peritumoral edema and/or normalizing blood brain barrier, and occasionally via reducing the tumor size. However, the effect against active cystic components has not been documented yet. Materials and Methods Between 2008 and 2018, 139 patients with primary or metastatic brain tumors were treated with bevacizumab (BEV) in our institution. The images and symptoms before and after administration of BEV were examined, and changes in size of cysts were evaluated as follows: CR (complete disappearance), PR (reduction by ≥50%), MR (reduction by ≥25%), SD (size change <25%), PD (increase by ≥25%). The effect of BEV on tumor itself was determined according to Response Assessment in Neuro‐Oncology criteria. Results Of the 139 patients, 21 (15.1%) had cystic components. The best responses of cysts to BEV treatment were as follows: CR 6, PR 7, MR 4, SD 4. The group of patients with progressively increasing cysts prior to BEV treatment had significant cyst size reduction compared to stable cyst size groups, at initial imaging after BEV (mean 62.6% vs 22.5%, P = .0055) and at best response timing (mean 76.3% vs 32.8%, P = .0050). Patients with cysts showed significant improvement in symptoms after the treatment with BEV compared to patients without cysts (P = .0033). However, response rate was not different between patients with or without cysts. Overall survival after starting BEV was not different between glioblastoma patients with or without cysts. Conclusion Bevacizumab is effective against progressively increasing cysts. Although cysts reduction effect and tumor response and/or overall survival are independent, BEV may be effective in patients who are symptomatic due to cyst enlargement.
Collapse
Affiliation(s)
- Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Manish Kolakshyapati
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoki Takano
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuki Imano
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shumpei Onishi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology & Neuro-oncology Program, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
25
|
Shimizu Y, Kajiyama H, Yoshida K, Tamauchi S, Nakanishi T, Kikkawa F. The usefulness of bevacizumab for relief from symptomatic malignant ascites in patients with heavily treated recurrent ovarian cancer. J Obstet Gynaecol Res 2019; 45:2435-2439. [PMID: 31468618 DOI: 10.1111/jog.14112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/14/2019] [Indexed: 02/02/2023]
Abstract
AIM Accumulation of ascites fluid is a major obstacle in the late phase of epithelial ovarian cancer. However, there is no consensus on a specific treatment for malignant ascites. The present study evaluated the clinical benefit of half-dose bevacizumab therapy (7.5 mg/kg every 3-4 weeks). METHODS This was a single-arm interventional study performed at Aichi Cancer Center Hospital. Four patients with platinum-resistant epithelial ovarian cancer and symptomatic malignant ascites were no longer considered candidates for standard chemotherapy. As a palliative approach, half-dose bevacizumab therapy (7.5 mg/kg every 3-4 weeks) was used with informed consent. The clinical data of these patients were retrospectively reviewed. RESULTS All patients had been heavily pretreated and showed progressive disease. Thus, standard chemotherapy was no longer feasible, and palliative paracentesis for malignant ascites was clinically needed. Among the four patients, three did not require additional paracentesis after bevacizumab therapy, and there were no adverse events. One patient needed paracentesis owing to lymphorrhea. CONCLUSION The use of bevacizumab therapy as a palliative approach for malignant ascites might be an option in patients with terminal-stage ovarian cancer. However, further evaluation is needed with regard to the possibility of severe side effects and medical expenses.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Gynecologic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Toru Nakanishi
- Department of Obstetrics and Gynecology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
26
|
Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc Natl Acad Sci U S A 2019; 116:2210-2219. [PMID: 30659155 DOI: 10.1073/pnas.1818357116] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In ovarian cancer patients, tumor fibrosis and angiotensin-driven fibrogenic signaling have been shown to inversely correlate with survival. We sought to enhance drug delivery and therapeutic efficacy by remodeling the dense extracellular matrix in two orthotopic human ovarian carcinoma xenograft models. We hypothesized that targeting the angiotensin signaling axis with losartan, an approved angiotensin system inhibitor, could reduce extracellular matrix content and the associated "solid stress," leading to better anticancer therapeutic effect. We report here four translatable findings: (i) losartan treatment enhances the efficacy of paclitaxel-a drug used for ovarian cancer treatment-via normalizing the tumor microenvironment, resulting in improved vessel perfusion and drug delivery; (ii) losartan depletes matrix via inducing antifibrotic miRNAs that should be tested as candidate biomarkers of response or resistance to chemotherapy; (iii) although losartan therapy alone does not reduce tumor burden, it reduces both the incidence and the amount of ascites formed; and (iv) our retrospective analysis revealed that patients receiving angiotensin system inhibitors concurrently with standard treatment for ovarian cancer exhibited 30 mo longer overall survival compared with patients on other antihypertensives. Our findings provide the rationale and supporting data for a clinical trial on combined losartan and chemotherapy in ovarian cancer patients.
Collapse
|
27
|
Miller D, Nevadunsky N. Palliative Care and Symptom Management for Women with Advanced Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:1087-1102. [DOI: 10.1016/j.hoc.2018.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Tew WP, Sill MW, Walker JL, Secord AA, Bonebrake AJ, Schilder JM, Stuckey A, Rice L, Tewari KS, Aghajanian CA. Randomized phase II trial of bevacizumab plus everolimus versus bevacizumab alone for recurrent or persistent ovarian, fallopian tube or peritoneal carcinoma: An NRG oncology/gynecologic oncology group study. Gynecol Oncol 2018; 151:257-263. [PMID: 30177462 PMCID: PMC6350932 DOI: 10.1016/j.ygyno.2018.08.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Bevacizumab (BV) monotherapy leads to compensatory upregulation of multiple signaling pathways, resulting in mTOR activation. We evaluated combining BV and everolimus (EV), an mTOR kinase inhibitor, to circumvent BV-resistance in women with recurrent or persistent ovarian, fallopian tube or primary peritoneal cancer (OC). PATIENTS AND METHODS Eligible OC patients had measurable (RECIST1.1) or detectable disease, 1-3 prior regimens, performance status (PS) 0-2, and no prior m-TOR inhibitor. All patients received BV 10 mg/kg IV every 2wks. Patients were randomized (1:1) to oral EV (10 mg daily) or placebo stratified by platinum-free interval (PFI), measurable disease and prior BV. Primary endpoint was progression-free survival (PFS); secondary endpoints included safety and response. RESULTS 150 patients were randomized to BV with (n = 75) and without (n = 75) EV. Arms were well-balanced for age (median 63: range 28-92), PS (0: 73%, 1-2: 27%), prior regimens (1: 37%, 2: 47%, 3: 16%), prior BV (11%), PFI (<6mos: 65%) and measurable disease (81%). The BV + EV vs BV median PFS was 5.9 vs 4.5 months (hazard ratio [HR] 0.95 (95% CI, 0.66-1.37, p = 0.39)). Median OS was 16.6 vs 17.3 months (HR 1.16 (95% CI, 0.72-1.87, p = 0.55). Objective measurable responses were higher with BV + EV (22% vs 12%). Study removal due to toxicity was higher with BV + EV (29% vs 12%). Toxicity (≥grade 3) from BV + EV were "other GI (mucositis)" (23 vs 1%) and "metabolic/nutrition" (19 vs. 7%); common ≥ grade 2 toxicities with BV + EV were cytopenia, nausea, fatigue and rash. CONCLUSION The combination regimen (BV + EV) did not significantly reduce the hazard of progression or death relative to BV and was associated with higher rates of adverse events and study discontinuation when compared to BV alone.
Collapse
Affiliation(s)
- William P Tew
- Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States of America.
| | - Michael W Sill
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park, Buffalo, NY 14263, United States of America.
| | - Joan L Walker
- Department of Gynecologic Oncology, The Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America.
| | - Angeles Alvarez Secord
- Dept. of Obstetrics/Gynecology, Division of Gynecology Oncology, Duke University Medical Center, Durham, NC 27710, United States of America.
| | - Albert J Bonebrake
- Cancer Research for the Ozarks-Cox Health, Ferrell_Duncan Clinic GYN-ONC, Springfield, MO 65807, United States of America.
| | - Jeanne M Schilder
- Indiana University Medical Center, Cancer Pavilion - Section of GYN Oncology, Indianapolis, IN 46202, United States of America.
| | - Ashley Stuckey
- Women and Infants Hospital, Program in Women's Oncology, Providence, RI 02905, United States of America.
| | - Laurel Rice
- University of Wisconsin, Obstetrics & Gynecology, Division of Gyn/Oncology, Madison, WI 53792, United States of America.
| | - Krishnansu S Tewari
- The Division of Gynecologic Oncology, University of California, Irvine Medical Center, Orange, CA 92868, United States of America.
| | - Carol A Aghajanian
- Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States of America.
| |
Collapse
|
29
|
Dei Cas M, Ghidoni R. Cancer Prevention and Therapy with Polyphenols: Sphingolipid-Mediated Mechanisms. Nutrients 2018; 10:nu10070940. [PMID: 30037082 PMCID: PMC6073226 DOI: 10.3390/nu10070940] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022] Open
Abstract
Polyphenols, chemically characterized by a polyhydroxylated phenolic structure, are well known for their widespread pharmacological properties: anti-inflammatory, antibiotic, antiseptic, antitumor, antiallergic, cardioprotective and others. Their distribution in food products is also extensive especially in plant foods such as vegetables, cereals, legumes, fruits, nuts and certain beverages. The latest scientific literature outlines a resilient interconnection between cancer modulation and dietary polyphenols by sphingolipid-mediated mechanisms, usually correlated with a modification of their metabolism. We aim to extensively survey this relationship to show how it could be advantageous in cancer treatment or prevention by nutrients. From this analysis it emerges that a combination of classical chemotherapy with nutrients and especially with polyphenols dietary sources may improve efficacy and decreases negative side effects of the antineoplastic drug. In this multifaceted scenario, sphingolipids play a pivotal role as bioactive molecules, emerging as the mediators of cell proliferation in cancer and modulator of chemotherapeutics.
Collapse
Affiliation(s)
- Michele Dei Cas
- Department of Health Sciences, University of Milan, 20142 Milan, Italy.
| | - Riccardo Ghidoni
- Department of Health Sciences, University of Milan, 20142 Milan, Italy.
| |
Collapse
|
30
|
Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat Commun 2018; 9:2270. [PMID: 29891938 PMCID: PMC5995921 DOI: 10.1038/s41467-018-04695-7] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/18/2018] [Indexed: 02/08/2023] Open
Abstract
The limitations of current anti-angiogenic therapies necessitate other targets with complimentary mechanisms. Here, we show for the first time that soluble E-cadherin (sE-cad) (an 80-kDa soluble form), which is highly expressed in the malignant ascites of ovarian cancer patients, is a potent inducer of angiogenesis. In addition to ectodomain shedding, we provide further evidence that sE-cad is abundantly released in the form of exosomes. Mechanistically, sE-cad-positive exosomes heterodimerize with VE-cadherin on endothelial cells and transduce a novel sequential activation of β-catenin and NFκB signaling. In vivo and clinical data prove the relevance of sE-cad-positive exosomes for malignant ascites formation and widespread peritoneal dissemination. These data advance our understanding of the molecular regulation of angiogenesis in ovarian cancer and support the therapeutic potential of targeting sE-cad. The exosomal release of sE-cad, which represents a common route for externalization in ovarian cancer, could potentially be biomarkers for diagnosis and prognosis. A soluble form E-cadherin is highly expressed in ovarian cancer. Here, the authors show that soluble E-cadherin is released by ovarian cancer cells packaged in exosomes and promotes tumor angiogenesis through β-catenin and NFkB signaling activation.
Collapse
|
31
|
Maki H, Nannya Y, Imai Y, Yamaguchi S, Kamikubo Y, Ichikawa M, Nakamura F, Kurokawa M. Nonmyelomatous Ascites Resulting from the Increased Secretion of Vascular Endothelial Growth Factor in Multiple Myeloma. Intern Med 2018; 57:725-727. [PMID: 29151500 PMCID: PMC5874348 DOI: 10.2169/internalmedicine.8886-17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ascites is a rare complication of multiple myeloma (MM); in most cases, the direct invasion of myeloma cells to the peritoneal cavity has been assumed to be the etiology because the effusion is usually exudative and contains a high proportion of myeloma cells. We herein report a case of MM with massive ascites containing only a small amount of myeloma cells. Instead, high levels of serum and ascitic vascular endothelial growth factor were detected. This was suggested to be a potential mechanism underlying the development of ascites.
Collapse
Affiliation(s)
- Hiroaki Maki
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yasuhito Nannya
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yoichi Imai
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Satoko Yamaguchi
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yasuhiko Kamikubo
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Motoshi Ichikawa
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Fumihiko Nakamura
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Mineo Kurokawa
- Department of Hematology & Oncology, Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
32
|
Chechlinska M, Kaminska J, Markowska J, Kramar A, Steffen J. Peritoneal Fluid Cytokines and the Differential Diagnosis of Benign and Malignant Ovarian Tumors and Residual/Recurrent Disease Examination. Int J Biol Markers 2018; 22:172-80. [DOI: 10.1177/172460080702200302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study aimed to assess the potential value of peritoneal fluid cytokine examination for the differential diagnosis of ovarian tumors and for evaluating residual or recurrent disease after treatment. The cytokines that are commonly elevated in ovarian cancer, VEGF, IL-6, bFGF, IL-8 and M-CSF, and a reference ovarian tumor marker, CA 125, were measured in peritoneal fluids of 53 previously untreated patients with epithelial ovarian cancer, 18 ovarian cancer patients after surgical treatment and chemotherapy, and 17 patients with benign epithelial ovarian tumors. Non-parametric statistical analysis of data was performed. Ovarian cancer peritoneal fluids, as compared to peritoneal fluids of patients with benign ovarian tumors, contained significantly higher concentrations of IL-6, VEGF and CA 125, and significantly lower concentrations of bFGF and M-CSF, but only the levels of IL-6 and VEGF were significantly higher in peritoneal fluids of stage I and II ovarian cancer patients than of patients with benign ovarian conditions. IL-6 at the cutoff level of 400 pg/mL discriminated benign and malignant ovarian tumors with 92% sensitivity and 60% specificity, while VEGF at the cutoff of 400 pg/mL had 90% sensitivity and 80% specificity. At the cutoff level of 1200 pg/mL, IL-6 had 84% sensitivity and 87% specificity. A radical decrease in local cytokine and CA 125 levels in patients after treatment was independent of therapy outcome. IL-6 and VEGF measurements in peritoneal fluids might be useful for the differential diagnosis of malignant and benign ovarian conditions, but not for residual or recurrent disease examination.
Collapse
Affiliation(s)
- M. Chechlinska
- Department of Immunology, Cancer
Center and Institute of Oncology, Warsaw
| | - J. Kaminska
- Department of Tumor Markers, Cancer
Center and Institute of Oncology, Warsaw - Poland
| | - J. Markowska
- Oncology Division, University of
Medical Science, Poznan - Poland
| | - A. Kramar
- Department of Biostatistics, CRLC Val
d'Aurelle, Parc Euromédecine, Montpellier - France
| | - J. Steffen
- Department of Immunology, Cancer
Center and Institute of Oncology, Warsaw
| |
Collapse
|
33
|
Lu CS, Lin JK, Chen WS, Lin TC, Jiang JK, Yang SH, Wang HS, Chang SC, Lan YT, Lin CC, Lin HH, Teng HW. Intraperitoneal ziv-aflibercept effectively manages refractory ascites in colorectal cancer patients. Oncotarget 2017; 8:36707-36715. [PMID: 27888621 PMCID: PMC5482690 DOI: 10.18632/oncotarget.13543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/12/2016] [Indexed: 01/07/2023] Open
Abstract
Ascites related to metastatic colorectal cancer (mCRC) reduces patient survival and quality of life, and systemic chemotherapy is largely ineffective for managing ascites. Here, we examined the clinical efficacy of intraperitoneal (IP) ziv-aflibercept for managing refractory ascites in 15 mCRC patients who did not respond to standard chemotherapy. Fifty or 100 mg of ziv-aflibercept in 100 mL of saline solution were infused through a pigtail catheter and retained for 24 h. When the ascites drainage volumes were subsequently monitored, 73.3% of patients showed an objective response (OR) to IP ziv-aflibercept treatment. Patients with low Eastern Cooperative Oncology Group (ECOG) performance status or with serum ascites albumin gradients (SAAG) less than 1.1 g/dL had better responses to treatment, and 4 patients with SAAG less than 1.1 g/dL showed rapid objective responses (rOR). These findings indicate that intraperitoneal ziv-aflibercept therapy may be a highly effective means of treating refractory ascites in mCRC patients, and that SAAG may be predictive of a rapid response to this treatment.
Collapse
Affiliation(s)
- Chieh-Sheng Lu
- Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jen-Kou Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Wei-Shone Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Tzu-Chen Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Jeng-Kai Jiang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Shung-Haur Yang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Huann-Sheng Wang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Shih-Ching Chang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Yuan-Tzu Lan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Chun-Chi Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Hung-Hsin Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Colon and Rectum Surgery, Department of Surgery, Taipei Veterans General Hospital & National Yang-Ming University, Taipei, Taiwan
| | - Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
34
|
Iwamura T, Narumi H, Suzuki T, Yanai H, Mori K, Yamashita K, Tsushima Y, Asano T, Izawa A, Momen S, Nishimura K, Tsuchiyama H, Uchida M, Yamashita Y, Okano K, Taniguchi T. Novel pegylated interferon-β as strong suppressor of the malignant ascites in a peritoneal metastasis model of human cancer. Cancer Sci 2017; 108:581-589. [PMID: 28129467 PMCID: PMC5406538 DOI: 10.1111/cas.13176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/15/2017] [Accepted: 01/21/2017] [Indexed: 12/19/2022] Open
Abstract
Malignant ascites manifests as an end‐stage event during the progression of a number of cancers and lacks a generally accepted standard therapy. Interferon‐β (IFN‐β) has been used to treat several cancer indications; however, little is known about the efficacy of IFN‐β on malignant ascites. In the present study, we report on the development of a novel, engineered form of human and murine IFN‐β, each conjugated with a polyethylene glycol molecule (PEG‐hIFN‐β and PEG‐mIFN‐β, respectively). We provide evidence that these IFN‐β molecules retain anti‐viral potency comparable to unmodified IFN‐β in vitro and manifested improved pharmacokinetics in vivo. Interestingly, PEG‐mIFN‐β significantly inhibited the accumulation of ascites fluid and vascular permeability of the peritoneal membrane in models of ovarian cancer and gastric cancer cell xenograft mice. We further show that PEG‐hIFN‐β directly suppresses VEGF165‐induced hyperpermeability in a monolayer of human vascular endothelial cells and that PEG‐mIFN‐β enhanced gene expression for a number of cell adhesion related molecules in mouse vascular endothelial cells. Taken together, these findings unveil a hitherto unrecognized potential of IFN‐β in maintaining vascular integrity, and provide proof‐of‐mechanism for a novel and long‐acting pegylated hIFN‐β for the therapeutic treatment of malignant ascites.
Collapse
Affiliation(s)
- Tomokatsu Iwamura
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hideki Narumi
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tomohiko Suzuki
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hideyuki Yanai
- Department of Molecular Immunology, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo, Japan
| | - Katsuyuki Mori
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Koji Yamashita
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Yoshiaki Tsushima
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tomomi Asano
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Akiko Izawa
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Shinobu Momen
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Kazumi Nishimura
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hiromi Tsuchiyama
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Masashi Uchida
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Yuji Yamashita
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Kiyoshi Okano
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tadatsugu Taniguchi
- Department of Molecular Immunology, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo, Japan
| |
Collapse
|
35
|
Bai L, Wang F, Li ZZ, Ren C, Zhang DS, Zhao Q, Lu YX, Wang DS, Ju HQ, Qiu MZ, Wang ZQ, Wang FH, Xu RH. Chemotherapy plus bevacizumab versus chemotherapy plus cetuximab as first-line treatment for patients with metastatic colorectal cancer: Results of a registry-based cohort analysis. Medicine (Baltimore) 2016; 95:e4531. [PMID: 28002313 PMCID: PMC5181797 DOI: 10.1097/md.0000000000004531] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The present observational cohort study was designed to elucidate the efficacy and safety profile of bevacizumab or cetuximab with chemotherapy as the first-line treatment in Chinese patients with metastatic colorectal cancer (mCRC). Clinical data were collected from a single-center registry study where mCRC patients received first-line fluoropyrimidine-based chemotherapy combined with either bevacizumab (188 patients with KRAS wild-type or mutated tumors) or cetuximab (101 patients with KRAS wild-type tumors) between January 2009 and December 2013. The Kaplan-Meier method was used for survival analysis. Cox proportional hazards model was used for estimating the prognostic and predictive values of clinicopathological characteristics. No statistically significant difference was observed between the bevacizumab and cetuximab groups in terms of median progression-free survival (PFS) (10.6 vs 8.7 months, P = 0.317), median overall survival (OS) (27.7 vs 28.3 months, P = 0.525), or overall response rate (43.1% vs 53.5%, P = 0.108). For the subset of patients with peritoneal dissemination, bevacizumab-based triplet appears to be superior to cetuximab-based triplet as measured by PFS (9.6 vs 6.1 months) and OS (26.3 vs 12.7 months), but not for patients without peritoneal dissemination (PFS, 10.6 vs 9.1 months; OS, 27.9 vs 30.7 months) (all unadjusted and adjusted interaction P < 0.05). Our study suggests that bevacizumab- or cetuximab-based regimens have similar effectiveness as first-line treatment of mCRC in Chinese population. Patients with peritoneal dissemination were likely to gain more benefit from bevacizumab than cetuximab treatment. Future prospective studies are required to further confirm these results.
Collapse
Affiliation(s)
- Long Bai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Feng Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Zhe-zhen Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Chao Ren
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Dong-sheng Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Qi Zhao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yun-xin Lu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - De-shen Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Huai-qiang Ju
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Miao-zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Zhi-qiang Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Feng-hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Rui-hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou
| |
Collapse
|
36
|
Tino AB, Chitcholtan K, Sykes PH, Garrill A. Resveratrol and acetyl-resveratrol modulate activity of VEGF and IL-8 in ovarian cancer cell aggregates via attenuation of the NF-κB protein. J Ovarian Res 2016; 9:84. [PMID: 27906095 PMCID: PMC5134119 DOI: 10.1186/s13048-016-0293-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/26/2016] [Indexed: 12/14/2022] Open
Abstract
Background Key features of advanced ovarian cancer include metastasis via cell clusters in the abdominal cavity and increased chemoresistance. Resveratrol and derivatives of resveratrol have been shown to have antitumour properties. The purpose of this study was to investigate the effect of resveratrol and acetyl-resveratrol on 3D cell aggregates of ovarian cancer, and establish if NF-κB signalling may be a potential target. Methods Poly-HEMA coated wells were used to produce 3D aggregates of two ovarian cancer cell lines, SKOV-3 and OVCAR-5. The aggregates were exposed to 10, 20 or 30 μM resveratrol or acetyl-resveratrol for 2, 4 or 6 days. Cell growth and metabolism were measured then ELISA, western blot and immunofluorescence were utilised to evaluate VEGF, IL-8 and NF-κB levels. Results Resveratrol and acetyl-resveratrol reduced cell growth and metabolism of SKOV-3 aggregates in a dose- and time-dependent manner. After 6 days all three doses of both compounds inhibited cell growth. This growth inhibition correlated with the attenuated secretion of VEGF and a decrease of NF-κB protein levels. Conversely, the secretion of IL-8 increased with treatment. The effects of the compounds were limited in OVCAR-5 cell clusters. Conclusions The results suggest that resveratrol and its derivative acetyl-resveratrol may inhibit in vitro 3D cell growth of certain subtypes of ovarian cancer, and growth restriction may be associated with the secretion of VEGF under the control of the NF-κB protein.
Collapse
Affiliation(s)
- Alexandria B Tino
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Kenny Chitcholtan
- Obstetrics and Gynaecology Department Christchurch Women's Hospital, Private Bag 4711, Christchurch, 8140, New Zealand.
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8140, New Zealand
| |
Collapse
|
37
|
Yin T, Wang G, He S, Shen G, Su C, Zhang Y, Wei X, Ye T, Li L, Yang S, Li D, Guo F, Mo Z, Wan Y, Ai P, Zhou X, Liu Y, Wang Y, Wei Y. Malignant Pleural Effusion and ascites Induce Epithelial-Mesenchymal Transition and Cancer Stem-like Cell Properties via the Vascular Endothelial Growth Factor (VEGF)/Phosphatidylinositol 3-Kinase (PI3K)/Akt/Mechanistic Target of Rapamycin (mTOR) Pathway. J Biol Chem 2016; 291:26750-26761. [PMID: 27756837 DOI: 10.1074/jbc.m116.753236] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/13/2016] [Indexed: 02/05/2023] Open
Abstract
Malignant pleural effusion (PE) and ascites, common clinical manifestations in advanced cancer patients, are associated with a poor prognosis. However, the biological characteristics of malignant PE and ascites are not clarified. Here we report that malignant PE and ascites can induce a frequent epithelial-mesenchymal transition program and endow tumor cells with stem cell properties with high efficiency, which promotes tumor growth, chemoresistance, and immune evasion. We determine that this epithelial-mesenchymal transition process is mainly dependent on VEGF, one initiator of the PI3K/Akt/mechanistic target of rapamycin (mTOR) pathway. From the clinical observation, we define a therapeutic option with VEGF antibody for malignant PE and ascites. Taken together, our findings clarify a novel biological characteristic of malignant PE and ascites in cancer progression and provide a promising and available strategy for cancer patients with recurrent/refractory malignant PE and ascites.
Collapse
Affiliation(s)
- Tao Yin
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Guoping Wang
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Sisi He
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Guobo Shen
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Chao Su
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yan Zhang
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xiawei Wei
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Tinghong Ye
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Ling Li
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Shengyong Yang
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Dan Li
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Fuchun Guo
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zeming Mo
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yang Wan
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Ping Ai
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xiaojuan Zhou
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yantong Liu
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yongsheng Wang
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yuquan Wei
- From the Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| |
Collapse
|
38
|
Chia PL, Russell PA, Scott AM, John T. Targeting the vasculature: anti-angiogenic agents for malignant mesothelioma. Expert Rev Anticancer Ther 2016; 16:1235-1245. [DOI: 10.1080/14737140.2016.1244008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Puey Ling Chia
- Department of Medical Oncology, Austin Health, Melbourne, Australia
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
| | - Prudence A. Russell
- Department of Anatomical Pathology, St. Vincent’s Hospital, University of Melbourne, Melbourne, Australia
| | - Andrew M Scott
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
- Faculty of Medicine, University of Melbourne, Melbourne, Australia
| | - Thomas John
- Department of Medical Oncology, Austin Health, Melbourne, Australia
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| |
Collapse
|
39
|
Solass W, Horvath P, Struller F, Königsrainer I, Beckert S, Königsrainer A, Weinreich FJ, Schenk M. Functional vascular anatomy of the peritoneum in health and disease. Pleura Peritoneum 2016; 1:145-158. [PMID: 30911618 PMCID: PMC6328070 DOI: 10.1515/pp-2016-0015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/30/2016] [Indexed: 01/28/2023] Open
Abstract
The peritoneum consists of a layer of mesothelial cells on a connective tissue base which is perfused with circulatory and lymphatic vessels. Total effective blood flow to the human peritoneum is estimated between 60 and 100 mL/min, representing 1-2 % of the cardiac outflow. The parietal peritoneum accounts for about 30 % of the peritoneal surface (anterior abdominal wall 4 %) and is vascularized from the circumflex, iliac, lumbar, intercostal, and epigastric arteries, giving rise to a quadrangular network of large, parallel blood vessels and their perpendicular offshoots. Parietal vessels drain into the inferior vena cava. The visceral peritoneum accounts for 70 % of the peritoneal surface and derives its blood supply from the three major arteries that supply the splanchnic organs, celiac and superior and inferior mesenteric. These vessels give rise to smaller arteries that anastomose extensively. The visceral peritoneum drains into the portal vein. Drugs absorbed are subject to first-pass hepatic metabolism. Peritoneal inflammation and cancer invasion induce neoangiogenesis, leading to the development of an important microvascular network. Anatomy of neovessels is abnormal and characterized by large size, varying diameter, convolution and blood extravasation. Neovessels have a defective ultrastructure: formation of large "mother vessels" requires degradation of venular and capillary basement membranes. Mother vessels give birth to numerous "daughter vessels". Diffuse neoangiogenesis can be observed before appearance of macroscopic peritoneal metastasis. Multiplication of the peritoneal capillary surface by neoangiogenesis surface increases the part of cardiac outflow directed to the peritoneum.
Collapse
Affiliation(s)
- Wiebke Solass
- Institute of Pathology, Medical School Hannover, Hannover, Germany
| | - Philipp Horvath
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Florian Struller
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Ingmar Königsrainer
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Stefan Beckert
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Frank-Jürgen Weinreich
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| | - Martin Schenk
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-Universitat Tubingen Medizinische Fakultat, Tuebingen, Germany
| |
Collapse
|
40
|
Vascular endothelial growth factor expression correlates with serum CA125 and represents a useful tool in prediction of refractoriness to platinum-based chemotherapy and ascites formation in epithelial ovarian cancer. Oncotarget 2016; 6:28491-501. [PMID: 26143638 PMCID: PMC4695074 DOI: 10.18632/oncotarget.4427] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 06/05/2015] [Indexed: 12/11/2022] Open
Abstract
There is an increasing need for the identification of novel biological markers and potential therapeutic targets in epithelial ovarian cancer (EOC). Given the critical role of growth factors in the biology of EOC, we aimed in the present study to evaluate the intratumoral expressions of vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) proteins and their clinical relevance in a cohort of 100 patients with EOC. All patients received platinum-based chemotherapy after surgery. A comparative immunohistochemical study of normal ovarian and EOC tissues showed that both growth factors were expressed at higher levels in tumor samples. In our statistical analysis, while no association existed between the FGF expression status and the clinicopathological characteristics of patients, intratumoral VEGF was identified as a potential biomarker for the prediction of ascites formation. In addition, the expression status of VEGF appeared to independently predict overall survival and response to chemotherapy. Furthermore, a direct association was demonstrated between the pre-treatment VEGF expression and serum CA125 after three cycles of chemotherapy. In sum, we report for the first time to our knowledge the correlation between intratumoral VEGF and serum CA125 in EOC. Our data also shows the prognostic value of VEGF expression in EOC. These results suggest the potential value of intratumoral VEGF in patient stratification. Dual inhibition of VEGF and CA125 might bring about a better outcome for patients with EOC.
Collapse
|
41
|
Tiper IV, Temkin SM, Spiegel S, Goldblum SE, Giuntoli RL, Oelke M, Schneck JP, Webb TJ. VEGF Potentiates GD3-Mediated Immunosuppression by Human Ovarian Cancer Cells. Clin Cancer Res 2016; 22:4249-58. [PMID: 27076627 DOI: 10.1158/1078-0432.ccr-15-2518] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/29/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Natural killer T (NKT) cells are important mediators of antitumor immune responses. We have previously shown that ovarian cancers shed the ganglioside GD3, which inhibits NKT-cell activation. Ovarian cancers also secrete high levels of VEGF. In this study, we sought to test the hypothesis that VEGF production by ovarian cancers suppresses NKT-cell-mediated antitumor responses. EXPERIMENTAL DESIGN To investigate the effects of VEGF on CD1d-mediated NKT-cell activation, a conditioned media model was established, wherein the supernatants from ovarian cancer cell lines (OV-CAR-3 and SK-OV-3) were used to treat CD1d-expressing antigen-presenting cells (APC) and cocultured with NKT hybridomas. Ovarian cancer-associated VEGF was inhibited by treatment with bevacizumab and genistein; conditioned medium was collected, and CD1d-mediated NKT-cell responses were assayed by ELISA. RESULTS Ovarian cancer tissue and ascites contain lymphocytic infiltrates, suggesting that immune cells traffic to tumors, but are then inhibited by immunosuppressive molecules within the tumor microenvironment. OV-CAR-3 and SK-OV-3 cell lines produce high levels of VEGF and GD3. Pretreatment of APCs with ascites or conditioned medium from OV-CAR-3 and SK-OV-3 blocked CD1d-mediated NKT-cell activation. Inhibition of VEGF resulted in a concomitant reduction in GD3 levels and restoration of NKT-cell responses. CONCLUSIONS We found that VEGF inhibition restores NKT-cell function in an in vitro ovarian cancer model. These studies suggest that the combination of immune modulation with antiangiogenic treatment has therapeutic potential in ovarian cancer. Clin Cancer Res; 22(16); 4249-58. ©2016 AACR.
Collapse
Affiliation(s)
- Irina V Tiper
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sarah M Temkin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland. Department of Gynecology and Obstetrics, The Kelly Gynecologic Oncology Service, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sarah Spiegel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Simeon E Goldblum
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Robert L Giuntoli
- Department of Gynecology and Obstetrics, The Kelly Gynecologic Oncology Service, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Mathias Oelke
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jonathan P Schneck
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
42
|
Sluiter NR, de Cuba EMV, Kwakman R, Meijerink WJHJ, Delis-van Diemen PM, Coupé VMH, Beliën JAM, Meijer GA, de Hingh IHJT, te Velde EA. Versican and vascular endothelial growth factor expression levels in peritoneal metastases from colorectal cancer are associated with survival after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Clin Exp Metastasis 2016; 33:297-307. [PMID: 26873137 PMCID: PMC4799792 DOI: 10.1007/s10585-016-9779-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022]
Abstract
Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) can increase survival of colorectal cancer (CRC) patients with peritoneal metastases (PM). This treatment is associated with high morbidity and mortality rates. Therefore, improvement of patient selection is necessary. Assuming that the clinical phenotype is dictated by biological mechanisms, biomarkers could play a crucial role in this process. Since it is unknown whether and to what extent angiogenesis influences the course of disease in patients with PM, we investigated the expression of two angiogenesis-related markers and their relation to overall survival (OS) in CRC patients after CRS and HIPEC. Clinicopathological data and tissue samples were collected from 65 CRC patients with isolated metastases to the peritoneum that underwent CRS and HIPEC. Whole tissue specimens from PM were evaluated for versican (VCAN) expression, VEGF expression and microvessel density (MVD) by immunohistochemistry. The relation between these markers and OS was assessed using univariate and multivariate analysis. Associations between VEGF expression, VCAN expression, MVD and clinicopathological data were tested. High stromal VCAN expression was associated with high MVD (p = 0.001), better resection outcome (p = 0.003) and high T-stage (p = 0.027). High epithelial VCAN expression was associated with MVD (p = 0.007) and a more complete resection (p < 0.001). In multivariate analysis, simplified peritoneal cancer index (p = 0.001), VEGF expression levels (p = 0.012), age (p = 0.030), epithelial VCAN expression levels (p = 0.042) and lymph node status (p = 0.053) were associated with OS. Concluding, VCAN and VEGF were associated with survival in CRC patients with PM after CRS and HIPEC. Independent validation in a well-defined patient cohort is required to confirm the putative prognostic role of these candidate biomarkers.
Collapse
Affiliation(s)
- N R Sluiter
- Section of Surgical Oncology and Digestive Surgery, Department of General Surgery, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - E M V de Cuba
- Section of Surgical Oncology and Digestive Surgery, Department of General Surgery, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Department of Pathology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Kwakman
- Section of Surgical Oncology and Digestive Surgery, Department of General Surgery, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - W J H J Meijerink
- Section of Surgical Oncology and Digestive Surgery, Department of General Surgery, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - P M Delis-van Diemen
- Department of Pathology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - V M H Coupé
- Department of Epidemiology and Biostatistics, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - J A M Beliën
- Department of Pathology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Department of Pathology, NKI-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - G A Meijer
- Department of Pathology, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Department of Pathology, NKI-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - I H J T de Hingh
- Department of Surgery, Catharina Hospital Eindhoven, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
| | - E A te Velde
- Section of Surgical Oncology and Digestive Surgery, Department of General Surgery, VU University Medical Centre, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Eto H, Hyodo F, Nakano K, Utsumi H. Selective Imaging of Malignant Ascites in a Mouse Model of Peritoneal Metastasis Using in Vivo Dynamic Nuclear Polarization-Magnetic Resonance Imaging. Anal Chem 2016; 88:2021-7. [PMID: 26796949 DOI: 10.1021/acs.analchem.5b04821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The presence of malignant ascites in advanced cancer patients is associated with both a poor prognosis and quality of life with a risk of abdominal infection and sepsis. Contemporary noninvasive visualization methods such as ultrasound, computed tomography, and magnetic resonance imaging (MRI) often struggle to differentiate malignant ascites from surrounding tissues. This study aimed to determine the utility of selective H2O imaging in the abdominal cavity with a free radical probe and deuterium oxide (D2O) contrast agent using in vivo dynamic nuclear polarization-MRI (DNP-MRI). Phantom imaging experiments established a linear relationship between H2O volume and image intensity using in vivo DNP-MRI. Similar results were obtained when the radical-D2O probe was used to determine selective and spatial information on H2O in vivo, modeled by the injection of saline into the abdominal cavity of mice. To demonstrate the utility of this method for disease, malignant ascites in peritoneal metastasis animal model was selected as one of the typical examples. In vivo DNP-MRI of peritoneal metastasis animal model was performed 7-21 days after intraperitoneal injection of luciferase, stably expressing the human pancreatic carcinoma (SUIT-2). The image intensity with increasing malignant ascites was significantly increased at days 7, 16, and 21. This increase corresponded to in vivo tumor progression, as measured by bioluminescent imaging. These results suggest that H2O signal enhancement in DNP-MRI using radical-D2O contrast is positively associated with the progression of dissemination and could be a useful biomarker for malignant ascites with cancer metastasis.
Collapse
Affiliation(s)
- Hinako Eto
- Innovation Center for Medical Redox Navigation, Kyushu University , 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Fuminori Hyodo
- Innovation Center for Medical Redox Navigation, Kyushu University , 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kenji Nakano
- Innovation Center for Medical Redox Navigation, Kyushu University , 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideo Utsumi
- Innovation Center for Medical Redox Navigation, Kyushu University , 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
44
|
de Cuba EMV, de Hingh IHJT, Sluiter NR, Kwakman R, Coupé VMH, Beliën JAM, Verwaal VJ, Meijerink WJHJ, Delis-van Diemen PM, Bonjer HJ, Meijer GA, Te Velde EA. Angiogenesis-Related Markers and Prognosis After Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Metastatic Colorectal Cancer. Ann Surg Oncol 2016; 23:1601-8. [PMID: 26727921 PMCID: PMC4819744 DOI: 10.1245/s10434-015-5023-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Patients presenting with peritoneal metastases (PM) of colorectal cancer (CRC) can be curatively treated with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Angiogenesis is under control of multiple molecules of which HIF1a, SDF1, CXCR4, and VEGF are key players. We investigated these angiogenesis-related markers and their prognostic value in patients with PM arising from CRC treated with CRS and HIPEC. PATIENTS AND METHODS Clinicopathological data and tissue specimens were collected in 2 tertiary referral centers from 52 patients who underwent treatment for isolated PM of CRC. Whole tissue specimens were subsequently analyzed for protein expression of HIF1a, SDF1, CXCR4, and VEGF by immunohistochemistry. Microvessel density (MVD) was analyzed by CD31 immunohistochemistry. The relationship between overall survival (OS) and protein expression as well as other clinicopathological characteristics was analyzed. RESULTS Univariate analysis showed that high peritoneal cancer index (PCI), resection with residual disease and high expression of VEGF were negatively correlated with OS after treatment with CRS and HIPEC (P < 0.01, P < 0.01, and P = 0.02, respectively). However, no association was found between the other markers and OS (P > 0.05). Multivariate analysis showed an independent association between OS and PCI, resection outcome and VEGF expression (multivariate HR: 6.1, 7.8 and 3.8, respectively, P ≤ 0.05). CONCLUSIONS An independent association was found between high VEGF expression levels and worse OS after CRS and HIPEC. The addition of VEGF expression to the routine clinicopathological workup could help to identify patients at risk for early treatment failure. Furthermore, VEGF may be a potential target for adjuvant treatment in these patients.
Collapse
Affiliation(s)
- E M V de Cuba
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - I H J T de Hingh
- Department of Surgery, Catharina Ziekenhuis Eindhoven, Eindhoven, The Netherlands
| | - N R Sluiter
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - R Kwakman
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - V M H Coupé
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - J A M Beliën
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - V J Verwaal
- Department of Surgery, Catharina Ziekenhuis Eindhoven, Eindhoven, The Netherlands
| | - W J H J Meijerink
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - P M Delis-van Diemen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - H J Bonjer
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - G A Meijer
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - E A Te Velde
- Department of Surgical Oncology, VU University Medical Center Amsterdam, Amsterdam, The Netherlands. .,Department of General Surgery, Section of Surgical Oncology and Digestive Surgery, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Razenberg LGEM, van Gestel YRBM, Lemmens VEPP, de Hingh IHJT, Creemers GJ. Bevacizumab in Addition to Palliative Chemotherapy for Patients With Peritoneal Carcinomatosis of Colorectal Origin: A Nationwide Population-Based Study. Clin Colorectal Cancer 2015; 15:e41-6. [PMID: 26762572 DOI: 10.1016/j.clcc.2015.12.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/27/2015] [Accepted: 12/09/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Most patients with colorectal cancer (CRC) presenting with peritoneal carcinomatosis (PC) rely on palliative systemic treatment options. However, data on the use and effect of systemic treatment strategies, including targeted agents for the palliative treatment of colorectal PC, are lacking. We conducted a nationwide population-based study with data from the period in which the targeted agent bevacizumab was introduced in the Netherlands. PATIENTS AND METHODS The present study included all patients diagnosed from 2007 to 2014 with synchronous PC from CRC treated with only palliative systemic therapy. We assessed the use of bevacizumab, the standard choice of targeted treatment, in addition to first-line chemotherapy. Multivariable logistic regression analyses were performed to calculate the predictors for the additional prescription of bevacizumab. Survival estimates were calculated, and multivariable Cox analyses were performed to estimate the hazard ratios (HRs) of death stratified by the treatment received. RESULTS A total of 1235 patients received palliative chemotherapy, of whom 436 also received bevacizumab (35%). Patients aged ≥ 75 years and patients with PC from colonic tumors were less likely to receive chemotherapy plus bevacizumab. The addition of bevacizumab to palliative chemotherapy was associated with an improved overall median survival of 7.5 versus 11 months in both patients with isolated PC and those with concomitant extraperitoneal metastases. The improvement remained after adjustment for patient and tumor characteristics (HR, 0.7; 95% confidence interval, 0.64-0.83). CONCLUSION The results of the present nationwide population-based study support the rationale for bevacizumab in addition to palliative chemotherapy for patients with PC of CRC and underline the need for ongoing efforts to precisely determine the role of targeted therapy in the treatment of PC.
Collapse
Affiliation(s)
- Lieke G E M Razenberg
- Department of Oncology, Catharina Hospital, Eindhoven, Netherlands; Department of Registry and Research, Netherlands Comprehensive Cancer Organization, Eindhoven, Netherlands.
| | - Yvette R B M van Gestel
- Department of Registry and Research, Netherlands Comprehensive Cancer Organization, Eindhoven, Netherlands
| | - Valery E P P Lemmens
- Department of Registry and Research, Netherlands Comprehensive Cancer Organization, Eindhoven, Netherlands; Department of Public Health, Erasmus Medical Center University Medical Centre, Rotterdam, Netherlands
| | | | | |
Collapse
|
46
|
VEGF-D as a marker in the aid of malignant metastatic pleural effusion diagnosis. Appl Immunohistochem Mol Morphol 2015; 23:209-14. [PMID: 25221955 DOI: 10.1097/pai.0000000000000079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The sensitivity in cytology diagnosis of malignant metastatic pleural effusion (MMPE) is insufficient nowadays due to the similarity of the reactive mesothelial cells and malignant cells. Vascular endothelial growth factor (VEGF) is one of the key factors in tumor lymphangiogenesis and metastasis. Therefore, the aim of this study was to evaluate the value of VEGF and its homologs in the aid of MMPE diagnosis. METHODS A total of 217 pleural effusions samples were eligible for analysis. Among them, 81 malignant and 22 benign cases were made into the cell blocks for the immunocytochemical (ICC) staining of VEGF-A, VEGF-C, VEGF-D, VEGFR-2, and VEGFR-3 expression. Another 114 samples (41 malignant and 73 benign cases) were subjected to the ELISA test for the protein level of VEGF-D. RESULTS In a total of 156 MMPE, only VEGF-D expression by ICC stain was significantly different between malignant (92.6%) and benign cases (9.1%) with P<0.001 in either nuclear or cytoplasmic staining. Only 6 malignant cases showed negative stain results. In addition, 3 of the 4 lung small cell carcinoma were immunoreactive for VEGF-D. However, some lymphocytes also showed nuclear staining pattern of VEGF-D. In contrast, the ELISA test for the VEGF-D protein levels failed to demonstrate the difference between malignant and benign pleural effusions. CONCLUSIONS Among VEGF homologs, MMPE from various kinds of tumor origin, VEGF-D showed 92.6% rate of positive expression. ICC stain of VEGF-D is a useful marker in the aid of MMPE diagnosis.
Collapse
|
47
|
Aggarwal S. Sustained Improvement in a Metastatic Colon Cancer Patient with FOLFIRI-Aflibercept after FOLFOX Failure. Case Rep Oncol 2015; 8:487-92. [PMID: 26668569 PMCID: PMC4677698 DOI: 10.1159/000441413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The present report illustrates a case of a 37-year-old Indian male patient diagnosed with adenocarcinoma of the sigmoid colon who underwent an anterior resection with total mesorectal excision surgery. He was administered adjuvant chemotherapy with 10 cycles of a FOLFOX-4 (folinic acid, fluorouracil and oxaliplatin) regimen but developed relapse. He was then put on a FOLFIRI (folinic acid, fluorouracil and irinotecan)-aflibercept (Zaltrap) regimen and received 12 cycles during the next 6 months. During the treatment period, a reduction in ascites along with a decline in serum carcinoembryonic antigen (CEA) level was observed, though the tumor size was unchanged. After completion of 12 cycles, the patient was asymptomatic but showed signs of progression in the form of increased CEA level. The FOLFIRI-aflibercept therapy was discontinued, and the patient was given supportive treatment, but he eventually died after another 6 months. The FOLFIRI-aflibercept treatment provided a progression-free survival of 6 months and an overall survival of 1 year to this patient, which corroborates the findings of the VELOUR trial.
Collapse
Affiliation(s)
- Shyam Aggarwal
- Department of Medical Oncology, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
48
|
Maeda H, Kobayashi M, Sakamoto J. Evaluation and treatment of malignant ascites secondary to gastric cancer. World J Gastroenterol 2015; 21:10936-10947. [PMID: 26494952 PMCID: PMC4607895 DOI: 10.3748/wjg.v21.i39.10936] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/26/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Malignant ascites affects approximately 10% of patients with gastric cancer (GC), and poses significant difficulties for both patients and clinicians. In addition to the dismal general condition of affected patients and the diversity of associated complications such as jaundice and ileus, problems in assessing scattered tumors have hampered the expansion of clinical trials for this condition. However, the accumulation of reported studies is starting to indicate that the weak response to treatment in GC patients with malignant ascites is more relevant to their poor prognosis rather than to the ascites volume at diagnosis. Therefore, precise assessment of initial state of ascites, repetitive evaluation of treatment efficacy, selection of suitable treatment, and swift transition to other treatment options as needed are paramount to maximizing patient benefit. Accurately determining ascites volume is the crucial first step in clinically treating a patient with malignant ascites. Ultrasonography is commonly used to identify the existence of ascites, and several methods have been proposed to estimate ascites volume. Reportedly, the sum of the depth of ascites at five points (named “five-point method”) on three panels of computed tomography images is well correlated to the actual ascites volume and/or abdominal girth. This method is already suited to repetitive assessment due to its convenience compared to the conventional volume rendering method. Meanwhile, a new concept, “Clinical Benefit Response in GC (CBR-GC)”, was recently introduced to measure the efficacy of chemotherapy for malignant ascites of GC. CBR-GC is a simple and reliable patient-oriented evaluation system based on changes in performance status and ascites, and is expected to become an important clinical endpoint in future clinical trials. The principal of treatment for GC patients with ascites is palliation and prevention of ascites-related symptoms. The treatment options are various, including a standard treatment based on the available guidelines, cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC), laparoscopic HIPEC alone, intravenous chemotherapy, intraperitoneal chemotherapy, and molecular targeting therapy. Although each treatment option is valid, further research is imperative to establish the optimal choice for each patient.
Collapse
|
49
|
Moughon DL, He H, Schokrpur S, Jiang ZK, Yaqoob M, David J, Lin C, Iruela-Arispe ML, Dorigo O, Wu L. Macrophage Blockade Using CSF1R Inhibitors Reverses the Vascular Leakage Underlying Malignant Ascites in Late-Stage Epithelial Ovarian Cancer. Cancer Res 2015; 75:4742-52. [PMID: 26471360 DOI: 10.1158/0008-5472.can-14-3373] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 08/23/2015] [Indexed: 11/16/2022]
Abstract
Malignant ascites is a common complication in the late stages of epithelial ovarian cancer (EOC) that greatly diminishes the quality of life of patients. Malignant ascites is a known consequence of vascular dysfunction, but current approved treatments are not effective in preventing fluid accumulation. In this study, we investigated an alternative strategy of targeting macrophage functions to reverse the vascular pathology of malignant ascites using fluid from human patients and an immunocompetent murine model (ID8) of EOC that mirrors human disease by developing progressive vascular disorganization and leakiness culminating in massive ascites. We demonstrate that the macrophage content in ascites fluid from human patients and the ID8 model directly correlates with vascular permeability. To further substantiate macrophages' role in the pathogenesis of malignant ascites, we blocked macrophage function in ID8 mice using a colony-stimulating factor 1 receptor kinase inhibitor (GW2580). Administration of GW2580 in the late stages of disease resulted in reduced infiltration of protumorigenic (M2) macrophages and dramatically decreased ascites volume. Moreover, the disorganized peritoneal vasculature became normalized and sera from GW2580-treated ascites protected against endothelial permeability. Therefore, our findings suggest that macrophage-targeted treatment may be a promising strategy toward a safe and effective means to control malignant ascites of EOC.
Collapse
Affiliation(s)
- Diana L Moughon
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Huanhuan He
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California
| | - Shiruyeh Schokrpur
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Ziyue Karen Jiang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Madeeha Yaqoob
- Department of Surgery and Cancer, Hammersmith hospital, Imperial College London, London, United Kingdom
| | - John David
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Los Angeles, California
| | - Crystal Lin
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, Los Angeles, California. Molecular Biology Institute, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | - Oliver Dorigo
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
50
|
Zhan N, Dong WG, Wang J. The clinical significance of vascular endothelial growth factor in malignant ascites. Tumour Biol 2015; 37:3719-25. [PMID: 26462841 DOI: 10.1007/s13277-015-4198-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/03/2014] [Indexed: 11/27/2022] Open
Abstract
Ascites can be caused by many kinds of diseases. Patients with undetermined ascites represent a diagnostic challenge. The aims of this study were to determine the diagnostic value of vascular endothelial growth factor (VEGF) in differentiation of malignant ascites from benign ascites and to investigate the clinical value of ascitic VEGF as an independent prognostic parameter. The study included 462 consecutive patients with malignant ascites and 550 patients with benign ascites, VEGF level in ascites were determined by a sandwich enzyme-linked immunosorbent assay. The survival rate was calculated by the Kaplan-Meier method and the log-rank test. Multivariate survival analysis was performed using the Cox hazards model. In our study, we found VEGF levels in malignant ascites (676.59 ± 303.86 pg/ml) were significantly higher than those in benign ascites (218.37 ± 98.15 pg/ml) (P < 0.001). Meanwhile, we also found that VEGF levels in malignant ascites from patients with ovarian cancer were higher than those with other cancers. Areas under the receiver operating characteristic (ROC) curves of ascitic VEGF was 0.940. At a cutoff value of 319.5 pg/ml, VEGF yielded a sensitivity of 89.2 % and a specificity of 88.4 %. Patients associated with the high-level VEGF value (≥613.38 pg/ml) in malignant ascites exhibited poor mean survival rates (8.3 ± 0.52 vs 15.11 ± 0.66 months, P < 0.001). In a multivariate Cox regression model, higher ascitic VEGF was an independent prognostic factor for overall survival. Planned subgroup analysis was performed for patients with tumor node metastasis (TNM) stage I. In the univariate analysis, only ascitic VEGF was associated with overall survival. VEGF was found to have a highly accurate sensitivity and specificity, suggesting that it could be considered as a new biomarker to differentiate malignant ascites from the benign one. The high level of VEGF value in malignant ascites may be used as an independent prognostic factor in patients with all stages of cancer.
Collapse
Affiliation(s)
- Na Zhan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Wei-Guo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Jing Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| |
Collapse
|