1
|
Chang YM, Huang WY, Yang SH, Jan CI, Nieh S, Lin YS, Chen SF, Lin YC. Interleukin-8/CXCR1 Signaling Contributes to the Progression of Pulmonary Adenocarcinoma Resulting in Malignant Pleural Effusion. Cells 2024; 13:968. [PMID: 38891100 PMCID: PMC11172099 DOI: 10.3390/cells13110968] [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: 03/18/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Pulmonary adenocarcinoma (PADC) treatment limited efficacy in preventing tumor progression, often resulting in malignant pleural effusion (MPE). MPE is filled with various mediators, especially interleukin-8 (IL-8). However, the role of IL-8 and its signaling mechanism within the fluid microenvironment (FME) implicated in tumor progression warrants further investigation. Primary cultured cells from samples of patients with MPE from PADC, along with a commonly utilized lung cancer cell line, were employed to examine the role of IL-8 and its receptor, CXCR1, through comparative analysis. Our study primarily assessed migration and invasion capabilities, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) properties. Additionally, IL-8 levels in MPE fluid versus serum, along with immunohistochemical expression of IL-8/CXCR1 signaling in tumor tissue and cell blocks were analyzed. IL-8/CXCR1 overexpression enhanced EMT and CSC properties. Furthermore, the immunocytochemical examination of 17 cell blocks from patients with PADC and MPE corroborated the significant correlation between upregulated IL-8 and CXCR1 expression and the co-expression of IL-8 and CXCR1 in MPE with distant metastasis. In summary, the IL-8/ CXCR1 axis in FME is pivotal to tumor promotion via paracrine and autocrine signaling. Our study provides a therapeutic avenue for improving the prognosis of PADC patients with MPE.
Collapse
Affiliation(s)
- Yi-Ming Chang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-M.C.); (S.-H.Y.)
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Wen-Yen Huang
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Shih-Hsien Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-M.C.); (S.-H.Y.)
- Office of General Affairs and Occupational Safety, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chia-Ing Jan
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Shin Nieh
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yaoh-Shiang Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Su-Feng Chen
- Department of Dentistry, School of Dentistry, China Medical University, Taichung 404333, Taiwan
| | - Yu-Chun Lin
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| |
Collapse
|
2
|
Accelerated Wound Healing and Keratinocyte Proliferation through PI3K/Akt/pS6 and VEGFR2 Signaling by Topical Use of Pleural Fluid. Cells 2022; 11:cells11050817. [PMID: 35269438 PMCID: PMC8909204 DOI: 10.3390/cells11050817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Impaired wound healing is an ongoing issue that cancer patients undergoing chemotherapy or radiotherapy face. Our previous study regarding lung-cancer-associated pleural fluid (LCPF) demonstrated its propensity to promote endothelial proliferation, migration, and angiogenesis, which are crucial features during cutaneous wound healing. Therefore, the current study aimed to investigate the effect of pleural fluid on cutaneous wound closure in vitro and in vivo using HaCaT keratinocytes and a full-thickness skin wound model, respectively. Both heart-failure-associated pleural fluid (HFPF) and LCPF were sequentially centrifuged and filtered to obtain a cell-free status. Treatment with HFPF and LCPF homogeneously induced HaCaT proliferation with cell cycle progression, migration, and MMP2 upregulation. Western blotting revealed increased PI3K/Akt phosphorylation and VEGFR2/VEGFA expression in HaCaT cells. When treated with the PI3K inhibitor, LCPF-induced keratinocyte proliferation was attenuated with decreased pS6 levels. By applying the VEGFR2 inhibitor, LCPF-induced keratinocyte proliferation was ameliorated by pS6 and MMP2 downregulation. The effect of LCPF-induced cell junction rearrangement was disrupted by co-treatment with a VEGFR2 inhibitor. Compared with a 0.9% saline dressing, LCPF significantly accelerated wound closure and re-epithelization when used as a dressing material in a full-thickness wound model. Histological analysis revealed increased neo-epidermis thickness and dermis collagen synthesis in the LCPF-treated group. Furthermore, LCPF treatment activated basal keratinocytes at the wound edge with the upregulation of Ki-67, VEGFA, and MMP2. Our preliminaries provided the benefit of wet dressing with pleural fluid to improve cutaneous wound closure through enhanced re-epithelization and disclosed future autologous application in cancer wound treatment.
Collapse
|
3
|
Plaksin SA, Farshatova LI. [Analysis of endothelial dysfunction in the context of pleural effusions of various origins using skin thermometry]. Khirurgiia (Mosk) 2020:59-63. [PMID: 33301255 DOI: 10.17116/hirurgia202012159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To analyze the mechanisms and severity of endothelial dysfunction in patients with pleural effusion of various origins using skin thermometry. MATERIAL AND METHODS We studied microcirculation in 135 patients with pleural effusions of various origins. Local skin thermometry and computer wavelet analysis were performed in 3-5 days after thoracoscopy with pleural biopsy using Microtest-100WF device with a temperature measuring resolution of 0.001°C. We estimated endothelial, myogenic and neurogenic indices using spectral analysis of skin temperature fluctuations in a range 0.0095-2 Hz. The control group comprised 40 healthy participants aged 23-36 years. RESULTS Three groups of patients were distinguished depending on the cause of effusion: malignant pleural effusions (n=65, 48.1%); inflammatory pleural effusions (para-pneumonic, post-traumatic, pancreatogenic, tuberculous) (n=58, 43%); transudates (hepatogenic, cardiogenic, and nephrogenic) (n=12, 8.9%). There were no significant differences in vascular tone in response to local heating of the second finger of the hand up to 40°C. Measurements were carried out within 10 min in all groups. Patients with diabetes mellitus and cardiovascular diseases (n=48, 35.6%) had significant decrease of thermal vasodilation index in endothelial range up to 2.93±1.77, in the control group - up to 4.21±2.40 (p=0.041). CONCLUSION Endothelial dysfunction is a universal non-specific aspect in pathogenesis of various diseases. This process is essential in pleural effusion. Local thermometry and computer wavelet analysis revealed no significant differences between patients with malignant, inflammatory and transudative pleural effusions. Pleural effusions in the context of cardiovascular pathology and/or diabetes mellitus occur due to impaired vasodilatation mechanisms in endothelial range.
Collapse
Affiliation(s)
- S A Plaksin
- Vagner Perm State Medical University, Perm, Russia
| | | |
Collapse
|
4
|
Guo M, Wu F, Hu G, Chen L, Xu J, Xu P, Wang X, Li Y, Liu S, Zhang S, Huang Q, Fan J, Lv Z, Zhou M, Duan L, Liao T, Yang G, Tang K, Liu B, Liao X, Tao X, Jin Y. Autologous tumor cell-derived microparticle-based targeted chemotherapy in lung cancer patients with malignant pleural effusion. Sci Transl Med 2020; 11:11/474/eaat5690. [PMID: 30626714 DOI: 10.1126/scitranslmed.aat5690] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/21/2018] [Accepted: 12/10/2018] [Indexed: 11/02/2022]
Abstract
Cell membrane-derived microparticles (MPs), the critical mediators of intercellular communication, have gained much interest for use as natural drug delivery systems. Here, we examined the therapeutic potential of tumor cell-derived MPs (TMPs) in the context of malignant pleural effusion (MPE). TMPs packaging the chemotherapeutic drug methotrexate (TMPs-MTX) markedly restricted MPE growth and provided a survival benefit in MPE models induced by murine Lewis lung carcinoma and colon adenocarcinoma cells. On the basis of the potential benefit and minimal toxicity of TMPs-MTX, we conducted a human study of intrapleural delivery of a single dose of autologous TMPs packaging methotrexate (ATMPs-MTX) to assess their safety, immunogenicity, and clinical activity. We report our findings on 11 advanced lung cancer patients with MPE. We found that manufacturing and infusing ATMPs-MTX were feasible and safe, without evidence of toxic effects of grade 3 or higher. Evaluation of the tumor microenvironment in MPE demonstrated notable reductions in tumor cells and CD163+ macrophages in MPE after ATMP-MTX infusion, which then translated into objective clinical responses. Moreover, ATMP-MTX treatment stimulated CD4+ T cells to release IL-2 and CD8+ cells to release IFN-γ. Our initial experience with ATMPs-MTX in advanced lung cancer with MPE suggests that ATMPs targeting malignant cells and the immunosuppressive microenvironment may be a promising therapeutic platform for treating malignancies.
Collapse
Affiliation(s)
- Mengfei Guo
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Wu
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guorong Hu
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lian Chen
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juanjuan Xu
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pingwei Xu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuan Wang
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yumei Li
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuqing Liu
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Zhang
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Huang
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinshuo Fan
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhilei Lv
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mei Zhou
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Limin Duan
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tingting Liao
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bifeng Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaofei Liao
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaonan Tao
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Jin
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
5
|
Bibby AC, Dorn P, Psallidas I, Porcel JM, Janssen J, Froudarakis M, Subotic D, Astoul P, Licht P, Schmid R, Scherpereel A, Rahman NM, Maskell NA, Cardillo G. ERS/EACTS statement on the management of malignant pleural effusions. Eur J Cardiothorac Surg 2019; 55:116-132. [PMID: 30060030 DOI: 10.1093/ejcts/ezy258] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 03/28/2018] [Indexed: 12/26/2022] Open
Abstract
Malignant pleural effusions (MPE) are a common pathology, treated by respiratory physicians and thoracic surgeons alike. In recent years, several well-designed randomized clinical trials have been published that have changed the landscape of MPE management. The European Respiratory Society (ERS) and the European Association for Cardio-Thoracic Surgery (EACTS) established a multidisciplinary collaboration of clinicians with expertise in the management of MPE with the aim of producing a comprehensive review of the scientific literature. Six areas of interest were identified, including the optimum management of symptomatic MPE, management of trapped lung in MPE, management of loculated MPE, prognostic factors in MPE, whether there is a role for oncological therapies prior to intervention for MPE and whether a histological diagnosis is always required in MPE. The literature revealed that talc pleurodesis and indwelling pleural catheters effectively manage the symptoms of MPE. There was limited evidence regarding the management of trapped lung or loculated MPE. The LENT score was identified as a validated tool for predicting survival in MPE, with Brims' prognostic score demonstrating utility in mesothelioma prognostication. There was no evidence to support the use of oncological therapies as an alternative to MPE drainage, and the literature supported the use of tissue biopsy as the gold standard for diagnosis and treatment planning.Management options for malignant pleural effusions have advanced over the past decade, with high-quality randomized trial evidence informing practice in many areas. However, uncertainties remain and further research is required http://ow.ly/rNt730jOxOS.
Collapse
Affiliation(s)
- Anna C Bibby
- Academic Respiratory Unit, University of Bristol Medical School Translational Health Sciences, Bristol, UK
- North Bristol Lung Centre, North Bristol NHS Trust, Bristol, UK
| | - Patrick Dorn
- Division of Thoracic Surgery, University Hospital Bern, Bern, Switzerland
| | | | - Jose M Porcel
- Pleural Medicine Unit, Arnau de Vilanova University Hospital, IRB Lleida, Lleida, Spain
| | - Julius Janssen
- Department of Pulmonary Diseases, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Marios Froudarakis
- Department of Respiratory Medicine, Medical School of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dragan Subotic
- Clinic for Thoracic Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Phillippe Astoul
- Department of Thoracic Oncology, Pleural Diseases and Interventional Pulmonology, Hospital North Aix-Marseille University, Marseille, France
| | - Peter Licht
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | - Ralph Schmid
- Division of Thoracic Surgery, University Hospital Bern, Bern, Switzerland
| | - Arnaud Scherpereel
- Pulmonary and Thoracic Oncology Department, Hospital of the University (CHU) of Lille, Lille, France
| | - Najib M Rahman
- Oxford Respiratory Trials Unit, University of Oxford, Oxford, UK
- Oxford Centre for Respiratory Medicine, University Hospitals, NHS Foundation Trust, Oxford, UK
| | - Nick A Maskell
- Academic Respiratory Unit, University of Bristol Medical School Translational Health Sciences, Bristol, UK
- North Bristol Lung Centre, North Bristol NHS Trust, Bristol, UK
- Task force chairperson
| | - Giuseppe Cardillo
- Task force chairperson
- Department of Thoracic Surgery, Carlo Forlanini Hospital, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| |
Collapse
|
6
|
Xie LX, Wang XG, You WJ, Ma XB, Wang YG, Liu TT, Jiang SJ. Predictors of survival in non-small cell lung cancer patients with pleural effusion undergoing thoracoscopy. Thorac Cancer 2019; 10:1412-1418. [PMID: 31094069 PMCID: PMC6558471 DOI: 10.1111/1759-7714.13086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/09/2019] [Accepted: 04/19/2019] [Indexed: 12/02/2022] Open
Affiliation(s)
- Li-Xu Xie
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xing-Guang Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wen-Jie You
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiao-Bin Ma
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yong-Gang Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Ting-Ting Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Shu-Juan Jiang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
7
|
Lim JU, Yeo CD, Kang HS, Park CK, Kim JS, Kim JW, Kim SJ, Lee SH. Elevated pretreatment platelet-to-lymphocyte ratio is associated with poor survival in stage IV non-small cell lung cancer with malignant pleural effusion. Sci Rep 2019; 9:4721. [PMID: 30886226 PMCID: PMC6423003 DOI: 10.1038/s41598-019-41289-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/04/2019] [Indexed: 02/07/2023] Open
Abstract
A higher platelet-to-lymphocyte ratio (PLR) has a clinical correlation with shorter survival in non-small cell lung cancer (NSCLC). The present study evaluated the association between the PLR and survival in patients with advanced NSCLC with malignant pleural effusion (MPE). Between January 2012 and July 2016, 237 patients with stage IV NSCLC were selected for evaluation. Receiver operating characteristic analysis was used to determine a cutoff for the PLR. Clinicopathological characteristics were compared between the high and low PLR groups, and the role of PLR as a predictive/prognostic maker was investigated. Among the 237 patients, 122 were assigned to the low PLR group and the other 115 to the high PLR group. According to multivariate analysis, male sex, not receiving active anticancer treatment, low hemoglobin level, low albumin level, high C-reactive protein level, and high PLR were identified as significant risk factors for shorter overall survival (OS) (p = 0.010, <0.001, 0.011, 0.004, 0.003, and <0.001, respectively). In the subgroup multivariate analysis of driver mutation-negative NSCLC, high Eastern Cooperative Oncology Group score, not receiving active anticancer treatment, low hemoglobin level, high C-reactive protein level, and high PLR were identified as significant risk factors for shorter OS (p = 0.047, <0.001, = 0.036, = 0.003, and <0.001, respectively). A high pretreatment PLR is independently associated with poor survival in stage IV NSCLC with MPE and in a subgroup of epidermal growth factor receptor and anaplastic lymphoma kinase wild-type NSCLC.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye Seon Kang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ju Sang Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Woo Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Haak Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Bibby AC, Dorn P, Psallidas I, Porcel JM, Janssen J, Froudarakis M, Subotic D, Astoul P, Licht P, Schmid R, Scherpereel A, Rahman NM, Cardillo G, Maskell NA. ERS/EACTS statement on the management of malignant pleural effusions. Eur Respir J 2018; 52:13993003.00349-2018. [DOI: 10.1183/13993003.00349-2018] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 03/28/2018] [Indexed: 02/07/2023]
Abstract
Malignant pleural effusions (MPE) are a common pathology, treated by respiratory physicians and thoracic surgeons alike. In recent years, several well-designed randomised clinical trials have been published that have changed the landscape of MPE management. The European Respiratory Society (ERS) and the European Association for Cardio-Thoracic Surgery (EACTS) established a multidisciplinary collaboration of clinicians with expertise in the management of MPE with the aim of producing a comprehensive review of the scientific literature.Six areas of interest were identified, including the optimum management of symptomatic MPE, management of trapped lung in MPE, management of loculated MPE, prognostic factors in MPE, whether there is a role for oncological therapies prior to intervention for MPE and whether a histological diagnosis is always required in MPE.The literature revealed that talc pleurodesis and indwelling pleural catheters effectively manage the symptoms of MPE. There was limited evidence regarding the management of trapped lung or loculated MPE. The LENT score was identified as a validated tool for predicting survival in MPE, with Brims' prognostic score demonstrating utility in mesothelioma prognostication. There was no evidence to support the use of oncological therapies as an alternative to MPE drainage, and the literature supported the use of tissue biopsy as the gold standard for diagnosis and treatment planning.
Collapse
|
9
|
Wang Z, Zheng Y, Fang Z. The clinical efficacy and safety of paclitaxel combined with avastin for NSCLC patients diagnosed with malignant pleural effusion. ACTA ACUST UNITED AC 2018; 64:230-233. [PMID: 29641777 DOI: 10.1590/1806-9282.64.03.230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/17/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The current study aimed to investigate the clinical efficacy of paclitaxel combined with avastin for non-small cell lung cancer (NSCLC) patients diagnosed with malignant pleural effusion (MPE). METHOD Total of 33 patients diagnosed with NSCLC as well as malignant pleural effusion were included. All of them received paclitaxel (175 mg/m2) and avastin (5 mg/kg). Clinical efficacy was evaluated using the total response rate, overall survival, progression-free survival and changes in MPE volume. Adverse events and rates of toxicities were examined as well. RESULTS The total response rate reached 77% while the overall survival and the median progression-free survival were respectively 22.2 months and 8.4 months. Toxicities of grade 3-4 consisted of neutropenia in 57% of patients, anemia in 17% of them, febrile neutropenia in 11%, as well as anorexia in 7%. No treatment-correlated deaths were found. CONCLUSION Paclitaxel combined with avastin decreased MPE volume and increased survival rate of NSCLC patients via inhibiting vascular endothelial growth factor expression.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Respiratory Medicine, Ningbo NO, 7 Hospital, China
| | - Yisheng Zheng
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical College of Fujian Medical University, Fuzhou General Hospital, China
| | - Zhenjian Fang
- Department of Respiratory Medicine, Fuding Hospital, China
| |
Collapse
|
10
|
Wu DW, Chang WA, Liu KT, Yen MC, Kuo PL. Vascular endothelial growth factor and protein level in pleural effusion for differentiating malignant from benign pleural effusion. Oncol Lett 2017; 14:3657-3662. [PMID: 28927127 DOI: 10.3892/ol.2017.6631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
Pleural effusion is associated with multiple benign and malignant conditions. Currently no biomarkers differentiate malignant pleural effusion (MPE) and benign pleural effusion (BPE) sensitively and specifically. The present study identified a novel combination of biomarkers in pleural effusion for differentiating MPE from BPE by enrolling 75 patients, 34 with BPE and 41 with MPE. The levels of lactate dehydrogenase, glucose, protein, and total cell, neutrophil, monocyte and lymphocyte counts in the pleural effusion were measured. The concentrations of interleukin (IL)-1β, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, tumor necrosis factor-α, interferon γ, transforming growth factor-β1, colony stimulating factor 2, monocyte chemoattractant protein-1 and vascular endothelial growth factor (VEGF) were detected using cytometric bead arrays. Protein and VEGF levels differed significantly between patients with BPE and those with MPE. The optimal cutoff value of VEGF and protein was 214 pg/ml and 3.35 g/dl respectively, according to the receiver operating characteristic curve. A combination of VEGF >214 pg/ml and protein >3.35 g/dl in pleural effusion presented a sensitivity of 92.6% and an accuracy of 78.6% for MPE, but was not associated with a decreased survival rate. These results suggested that this novel combination strategy may provide useful biomarkers for predicting MPE and facilitating early diagnosis.
Collapse
Affiliation(s)
- Da-Wei Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Kuan-Ting Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan, R.O.C
| |
Collapse
|
11
|
Mori R, Fujimoto D, Ito M, Tomii K. Bevacizumab for ramucirumab refractory malignant pleural effusion in non-small cell lung cancer: a case report and review of the literature. Oncotarget 2017; 8:48521-48524. [PMID: 28591698 PMCID: PMC5564666 DOI: 10.18632/oncotarget.17952] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/01/2017] [Indexed: 01/30/2023] Open
Abstract
Malignant pleural effusion (MPE) is a major problem associated with advanced non-small cell lung cancer for which an optimum treatment strategy has yet to be determined. Notably, vascular endothelial growth factor (VEGF) signaling has been found to influence MPE, and bevacizumab, a VEGF ligand inhibitor, can effectively control MPE. Ramucirumab, a human monoclonal antibody specific for VEGF receptor-2, has recently been approved for advanced non-small cell lung cancer. However, it remains unclear which of these agents more effectively control MPE.We describe a case of a 68-year-old man with advanced non-small cell lung cancer in whom ramucirumab plus docetaxel-refractory MPE was responsive to bevacizumab plus docetaxel combination therapy. The patient's MPE progressed after two cycles of ramucirumab plus docetaxel second-line chemotherapy. After switching to bevacizumab plus docetaxel, a computed tomography scan revealed a decreased MPE after two cycles of treatment.Bevacizumab may be more effective for treating MPE. However, further investigations are still warranted to determine the optimal VEGF-targeted agent for this condition.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents, Immunological/therapeutic use
- Bevacizumab/therapeutic use
- Carcinoma, Non-Small-Cell Lung/diagnostic imaging
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Drug Resistance, Neoplasm
- Humans
- Lung Neoplasms/diagnostic imaging
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Male
- Pleural Effusion, Malignant/pathology
- Tomography, X-Ray Computed
- Treatment Outcome
- Ramucirumab
Collapse
Affiliation(s)
- Ryobu Mori
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Daichi Fujimoto
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Munehiro Ito
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Hyogo, Japan
| |
Collapse
|
12
|
The Exploration of Peptide Biomarkers in Malignant Pleural Effusion of Lung Cancer Using Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry. DISEASE MARKERS 2017; 2017:3160426. [PMID: 28386154 PMCID: PMC5366757 DOI: 10.1155/2017/3160426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/15/2017] [Indexed: 11/19/2022]
Abstract
Background. Diagnoses of malignant pleural effusion (MPE) are a crucial problem in clinics. In our study, we compared the peptide profiles of MPE and tuberculosis pleural effusion (TPE) to investigate the value of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) in diagnosis of MPE. Material and Methods. The 46 MPE and 32 TPE were randomly assigned to training set and validation set. Peptides were isolated by weak cation exchange magnetic beads and peaks in the m/z range of 800–10000 Da were analyzed. Comparing the peptide profile between 30 MPE and 22 TPE samples in training set by ClinProTools software, we screened the specific biomarkers and established a MALDI-TOF-MS classification of MPE. Finally, the other 16 MPE and 10 TPE were included to verify the model. We additionally determined carcinoembryonic antigen (CEA) in MPE and TPE samples using electrochemiluminescent immunoassay method. Results. Five peptide peaks (917.37 Da, 4469.39 Da, 1466.5 Da, 4585.21 Da, and 3216.87 Da) were selected to separate MPE and TPE by MALDI-TOF-MS. The sensitivity, specificity, and accuracy of the classification were 93.75%, 100%, and 96.15%, respectively, after blinded test. The sensitivity of CEA was significantly lower than MALDI-TOF-MS classification (P = 0.035). Conclusions. The results indicate MALDI-TOF-MS is a potential method for diagnosing MPE.
Collapse
|
13
|
Dixit R, Agarwal KC, Gokhroo A, Patil CB, Meena M, Shah NS, Arora P. Diagnosis and management options in malignant pleural effusions. Lung India 2017; 34:160-166. [PMID: 28360465 PMCID: PMC5351359 DOI: 10.4103/0970-2113.201305] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant pleural effusion (MPE) denotes an advanced malignant disease process. Most of the MPE are metastatic involvement of the pleura from primary malignancy at lung, breast, and other body sites apart from lymphomas. The diagnosis of MPE has been traditionally made on cytological examination of pleural fluid and/or histological examination of pleural biopsy tissue that still remains the initial approach in these cases. There has been tremendous advancement in the diagnosis of MPE now a day with techniques i.e. characteristic Ultrasound and computed tomography features, image guided biopsies, fluorodeoxyglucose-positron emission tomography imaging, thoracoscopy with direct biopsy under vision, tumor marker studies and immunocytochemical analysis etc., that have made possible an early diagnosis of MPE. The management of MPE still remains a challenge to pulmonologist and oncologist. Despite having various modalities with better tolerance such as pleurodesis and indwelling pleural catheters etc., for long-term control, all the management approaches remain palliative to improve the quality of life and reduce symptoms. While choosing an appropriate management intervention, one should consider the clinical status of the patient, life expectancy, overall cost, availability and comparative institutional outcomes, etc.
Collapse
Affiliation(s)
- Ramakant Dixit
- Department of Respiratory Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - K C Agarwal
- Department of Respiratory Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - Archana Gokhroo
- Department of Internal Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - Chetan B Patil
- Department of Respiratory Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - Manoj Meena
- Department of Respiratory Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - Narender S Shah
- Department of Radiation Oncology, J.L.N. Medical College, Ajmer, Rajasthan, India
| | - Piyush Arora
- Department of Respiratory Medicine, J.L.N. Medical College, Ajmer, Rajasthan, India
| |
Collapse
|
14
|
Qi N, Li F, Li X, Kang H, Zhao H, Du N. Combination use of paclitaxel and avastin enhances treatment effect for the NSCLC patients with malignant pleural effusion. Medicine (Baltimore) 2016; 95:e5392. [PMID: 27893676 PMCID: PMC5134869 DOI: 10.1097/md.0000000000005392] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The current study is conducted to investigate efficacy of the chemotherapy drug paclitaxel in combination with Avastin (Roche Diagnostics GmbH., Mannheim, Germany) (antiangiogenic agent) in treatment of malignant pleural effusions (MPEs).Twenty-four patients with non-small cell lung cancer were randomly assigned for 2 treatment approaches. Ten patients received paclitaxel (175 mg/m) alone, and 14 patients took a combination therapy of paclitaxel and Avastin (5 mg/kg). Efficacy of the treatment approaches in the patients was validated with the change in the MPE volume. Pharmacokinetic (PK) profile and urinary excretion rate of paclitaxel were analyzed with serum vascular endothelial growth factor (VEGF) level, and adverse events were examined as well.The combination therapy reduced the MPE level with a successful rate of 29% and a survival rate of 25% over the single paclitaxel treatment in the study cohort (both P < 0.05). PKs for the combined treatment displayed a rapid distribution of the anticancer drug paclitaxel with an obvious increase in its elimination half-life in the pleural fluid (both P < 0.01). Mean residence time of paclitaxel increased in the presence of Avastin (P < 0.01). Serum VEGF levels significantly reduced in the Avastin-treated patients as compared to the paclitaxel-treated ones (P < 0.01). The urinary excretion rate was similar in the study cohort. Incidence of adverse events for the 2 treatment approaches was similar in the patients.Intervention of Avastin enhances potency of paclitaxel in treatment of MPEs with the increased survival rate of the patients through inhibiting VEGF production and prolonging time of ongoing interaction between the chemotherapy drug and the tumor tissues.
Collapse
Affiliation(s)
- Nan Qi
- Department of Oncology, The First Affiliated Hospital
| | - Fang Li
- Tumor Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaosong Li
- Department of Oncology, The First Affiliated Hospital
| | - Huanrong Kang
- Department of Oncology, The First Affiliated Hospital
| | - Hui Zhao
- Department of Oncology, The First Affiliated Hospital
| | - Nan Du
- Department of Oncology, The First Affiliated Hospital
| |
Collapse
|
15
|
Yang J, Zhu Y, Wu L, Zhu W, Zhang X, Yang Y, Xu C. Diagnostic and prognostic significance of receptor-binding cancer antigen expressed on SiSo cells in lung-cancer-associated pleural effusion. CLINICAL RESPIRATORY JOURNAL 2016; 12:279-284. [PMID: 27402224 DOI: 10.1111/crj.12527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/19/2016] [Accepted: 07/10/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This study aimed to evaluate the diagnostic and prognostic value of pleural effusion levels of soluble receptor-binding cancer antigen expressed on SiSo cells (sRCAS1) in lung cancer patients with malignant pleural effusion (MPE). METHODS Pleural effusion samples were collected from 78 patients with MPE, and from 48 patients with benign pleural effusion (BPE). Pleural effusion sRCAS1 concentrations were measured by enzyme-linked immunosorbent assay. RESULTS MPE has significantly higher sRCAS1 levels than that of BPE (P < .01). With a cutoff value of 18.7 U/mL, sRCAS1 showed a good diagnostic performance for MPE. Univariate and multivariate analysis indicated that elevated sRCAS1 levels were an independent predictor of overall survival (OS) and disease-free survival (DFS). Kaplan-Meier survival curves further confirmed that patients with high sRCAS1 have shorter DFS and OS (P = .026 and P = .032, respectively). CONCLUSION In conclusion, measurement of sRCAS1 might be a useful diagnostic and prognostic marker for MPE.
Collapse
Affiliation(s)
- Jian Yang
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Ying Zhu
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Liangquan Wu
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Wenyan Zhu
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Xiuwei Zhang
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Yang Yang
- Department of Respiratory Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Chunhua Xu
- Endoscopic Center of Nanjing Chest Hospital, Nanjing, Jiangsu, 210029, China.,Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing, Jiangsu, 210029, China
| |
Collapse
|
16
|
JIA LIN, DU YUN, LI TAO, LV YALEI, WANG YUDONG, ZHANG YAN, ZHOU XINLIANG, LIU WEI. Differential expression of vascular endothelial growth factor-A, -C and -D for the diagnosis and prognosis of cancer patients with malignant effusions. Oncol Lett 2015; 10:667-674. [PMID: 26622551 PMCID: PMC4508981 DOI: 10.3892/ol.2015.3305] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
Elevated levels of vascular endothelial growth factor (VEGF) contribute to angiogenesis and serous cavity effusions. The present study evaluated the diagnostic and prognostic values of VEGF-A, -C and -D proteins in the serum, supernatant fluid and exfoliated cells of cancer patients with malignant effusions compared with patients with benign effusions. An enzyme-linked immunosorbent assay was used to detect levels of VEGF-A, -C and -D proteins in the sera of 79 cases (30 lung cancer, 21 gastric cancer and 28 benign effusions) and the supernatant fluid of 96 cases (38 lung cancer, 30 gastric cancer, and 28 benign effusion). Immunocytochemistry detected the expression of VEGF-A, -C and -D proteins in effusion cells from 71 cases (34 lung cancer, 17 gastric cancer and 20 benign effusions). The data were further investigated to determine whether there was an association between VEGF subtype expression and clinicopathological characteristics and prognosis. The expression levels of VEGF-A in the supernatant fluid were increased in the lung and gastric cancer patient samples compared with the benign effusions (P<0.05). The VEGF-A level in the supernatant fluid was significantly increased compared with the corresponding sera of patients with malignant effusion (P<0.05). VEGF-A, -C and -D proteins in the exfoliated cells from primary lung or gastric cancer effusions were expressed at 52.94, 70.58 and 82.35%, respectively, whereas their expression was not detected in the exfoliated cells from benign effusion, with the exception of mesothelial cells. The levels of VEGF-A and VEGF-C in the supernatant fluid levels and the cell levels of VEGF-A were inversely associated with age; in addition, VEGF-A levels in the supernatant fluid were associated with malignant and bloody effusion, and only cavity metastasis (P<0.05). Survival analysis demonstrated a relatively reduced survival time for patients with VEGF-A levels of >406.19 pg/ml in the supernatant fluid compared with patients with VEGF-A levels of ≤406.19 pg/ml (P=0.066). Serum VEGF-A, -C and -D levels exhibited no evident clinical significance in the diagnosis and prognosis of serous cavity effusions. VEGF-A in the supernatant fluid merits further study as a tumor marker in the clinical setting to discriminate benign from malignant effusions, while cellular VEGF-C and -D may contribute to the formation of malignant effusions.
Collapse
Affiliation(s)
- LIN JIA
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - YUN DU
- Hebei Province China-Japan Friendship Center for Cancer Detection, Shijiazhuang, Hebei 050017, P.R. China
| | - TAO LI
- Department of Epidemiology and Health Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - YALEI LV
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - YUDONG WANG
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - YAN ZHANG
- Hebei Province China-Japan Friendship Center for Cancer Detection, Shijiazhuang, Hebei 050017, P.R. China
| | - XINLIANG ZHOU
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - WEI LIU
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
17
|
Spella M, Giannou AD, Stathopoulos GT. Switching off malignant pleural effusion formation-fantasy or future? J Thorac Dis 2015; 7:1009-20. [PMID: 26150914 DOI: 10.3978/j.issn.2072-1439.2015.05.20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Abstract
Malignant pleural effusion (MPE) is common and difficult to treat. In the vast majority of patients the presence of MPE heralds incurable disease, associated with poor quality of life, morbidity and mortality. Current therapeutic approaches are inefficient and merely offer palliation of associated symptoms. Recent scientific progress has shed light in the biologic processes governing the mechanisms behind the pathobiology of MPE. Pleural based tumors interfere with pleural fluid drainage, as well as the host vasculature and immune system, resulting in decreased fluid absorption and increased pleural fluid production via enhanced plasma extravasation into the pleural space. In order to achieve this feat, pleural based tumors must elicit critical vasoactive events in the pleura, thus forming a favorable microenvironment for tumor dissemination and MPE development. Such properties involve specific transcriptional signaling cascades in addition to secretion of important mediators which attract and activate host cell populations which, in turn, impact tumor cell functions. The dissection of the biologic steps leading to MPE formation provides novel therapeutic targets and recent research findings provide encouraging results towards future therapeutic innovations in MPE management.
Collapse
Affiliation(s)
- Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, 26504, Greece
| | - Anastasios D Giannou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, 26504, Greece
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, 26504, Greece
| |
Collapse
|
18
|
Zhang Y, Yu LK, Lu GJ, Xia N, Xie HY, Hu W, Hao KK, Xu CH, Qian Q. Prognostic values of VEGF and endostatin with malignant pleural effusions in patients with lung cancer. Asian Pac J Cancer Prev 2015; 15:8435-40. [PMID: 25339042 DOI: 10.7314/apjcp.2014.15.19.8435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AIMS Angiogenesis is important in malignant pleural effusion (MPE) formation and it is regulated by a number of pro- and anti-angiogenic cytokines. The purpose of this study was to evaluate the prognostic value of angiogenic factor vascular endothelial growth factor (VEGF) and angiogenesis inhibitor endostatin in lung cancer patients with MPE, and investigate the relationship between these two kinds of agent. METHODS Using enzyme-linked immunoadsorbent assay, the concentrations of VEGF and endostatin were measured in pleural effusions (PE) and serum from a total of 70 lung cancer patients with MPE and 20 patients with tuberculosis. RESULTS Compared to patients with tuberculosis, the levels of VEGF and endostatin in both PE and serum were significantly higher in patients with lung cancer. There were statistically significant correlations between VEGF levels in PE and serum (r=0.696, <0.001), endostatin levels in PE and serum (r=0.310, p=0.022), and VEGF and endostatin levels in PE (r=0.287, p=0.019). Cox multivariate analysis revealed that elevated pleural VEGF and endostatin levels and serum endostatin level were independent predictors of shorter overall survival. CONCLUSION Both pro- and anti-angiogenic factors are likely contributors to PE formation. Our results suggest that the levels of VEGF and endostatin in PE, together with endostatin in serum, may be potential prognostic parameters for lung cancer patients with MPE.
Collapse
Affiliation(s)
- Yu Zhang
- First Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing, Jiangsu Province, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Clinical implications of serum hypoxia inducible factor-1α and vascular endothelial growth factor in lung cancer. TUMORI JOURNAL 2015; 101:404-11. [PMID: 25983091 DOI: 10.5301/tj.5000320] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2015] [Indexed: 11/20/2022]
Abstract
AIMS AND BACKGROUND Hypoxia inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) have been deemed as key in angiogenesis of lung cancer. The aim of this study was to investigate diagnostic and prognostic values of HIF-1α and VEGF in patients with lung cancer. METHODS From May 1, 2011, to April 20, 2014, blood samples and/or pleural effusions were collected from 100 patients with lung cancer, 18 patients with tuberculosis, 47 patients with community-acquired pneumonia, and 29 healthy controls. The pretreatment levels of HIF-1α and VEGF were measured by enzyme-linked immunoassays. Patients with lung cancer were followed up during the period of this study and survival times were recorded for analysis. RESULTS We detected that the levels of serum and pleural HIF-1α in lung cancer were significantly higher than those in the tuberculosis population, and that the VEGF expressions were not significantly different between malignancy and benign diseases. An area under the curve of pleural HIF-1α (0.877 ± 0.053) showed a high ability to differentiate lung cancer from benign diseases. The significant negative predictors of survival in the univariate analysis were performance status (gt;1), no anticancer therapy, low serum albumin, advanced stage, and serum high level of VEGF (gt;324.17 pg/mL), while in the multivariate Cox regression analysis, only the pretreatment serum level of VEGF, stage, and anticancer therapy were identified as independent prognostic factors. CONCLUSIONS The overexpression of HIF-1α especially in pleural effusion may be an angiogenic factor for distinguishing malignancy from tuberculosis, and the pretreatment level of serum VEGF may be an independent predictor of survival.
Collapse
|
20
|
Wang Y, Yao Y, Liu H, Ma X, Lv T, Yuan D, Xiao X, Yin J, Song Y. Itraconazole can inhibit malignant pleural effusion by suppressing lymphangiogenesis in mice. Transl Lung Cancer Res 2015; 4:27-35. [PMID: 25806344 DOI: 10.3978/j.issn.2218-6751.2014.11.03] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 10/06/2014] [Indexed: 01/30/2023]
Abstract
BACKGROUND The presence of malignant pleural effusion (MPE) indicates a poor prognosis in patients with non-small cell lung cancer (NSCLC). Itraconazole has been identified as a potent inhibitor of endothelial cell proliferation that suppresses angiogenesis; however, its role in the suppression of lymphangiogenesis is still unclear. The aim of this study was to investigate the efficacy of itraconazole for MPE and the mechanism of lymphangiogenesis suppression. METHODS Lewis lung carcinoma (LLC) cells were injected into the mouse pleural cavity to establish the MPE mouse model, followed by randomization of the mice into three groups. Each mice was injected with either a high dose of itraconazole (25 mg/kg, H-ITCZ), a low dose of itraconazole (8 mg/kg, L-ITCZ), or 50 μL of hydroxypropyl-β-cyclodextrin (130 mg/mL, H-β-C) into the pleural cavity four times every 3 days. The MPE of the mice was collected and measured with a 1 mL syringe. The vascular endothelial growth factor-C (VEGF-C) expression level in the MPE was detected by enzyme-linked immunosorbent assay (ELISA), while the VEGF-C expression and lymphatic micro vessel density (LMVD) in the tumor tissue was observed by immunohistochemistry (IHC) staining. RESULTS The number of pleural tumor foci, the volume of pleural effusion, the LMVD and the VEGF-C expression levels in the tumor tissue were significantly reduced in the H-ITCZ-treated group. CONCLUSIONS Our results revealed that itraconazole may play an important role in the MPE mice by suppressing lymphangiogenesis, which demonstrated the usefulness of itraconazole in the treatment of MPE.
Collapse
Affiliation(s)
- Yunfen Wang
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Yanwen Yao
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Hongbin Liu
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Xingqun Ma
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Tangfeng Lv
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Dongmei Yuan
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Xinwu Xiao
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Jie Yin
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| | - Yong Song
- 1 Department of Respiratory Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing 210002, China ; 2 Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China ; 3 Department of Medical Oncology, 81 Hospital of PLA, Nanjing 210002, China
| |
Collapse
|
21
|
Xu CH, Cao L, Zhang XW, Yan J, Yu LK. Prognostic value of soluble H7-B4 in pleural effusion associated with lung cancer. Tumour Biol 2015; 36:4397-403. [PMID: 25636447 DOI: 10.1007/s13277-015-3079-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/08/2015] [Indexed: 11/29/2022] Open
Abstract
B7-H4, a member of the inhibitory B7 family, can restrain T cell proliferation, activation, and cytokine secretion and may be involved in immune evasion in cancer patients. This study aimed to evaluate the diagnostic and prognostic value of pleural effusion levels of soluble B7-H4 (sB7-H4) in lung cancer patients with malignant pleural effusion (MPE). Pleural effusion samples were collected from 98 lung cancer patients with malignant effusion and from 60 patients with nonmalignant pleural effusion. Pleural effusion concentrations of sB7-H4 were measured using sandwich enzyme-linked immunosorbent assay. Malignant effusion exhibited higher sB7-H4 levels than those in nonmalignant effusion (P < 0.01). Lung cancer patients with pleural effusion sB7-H4 levels below 35.8 ng/ml had a longer overall survival than those with higher levels (P < 0.05). By multivariate analysis, pleural effusion sB7-H4 was an independent prognostic factor in patients with MPE. In conclusion, measurement of sB7-H4 might be a useful diagnostic and prognostic value for MPE patients.
Collapse
Affiliation(s)
- Chun-Hua Xu
- Department of Respiratory Medicine, Nanjing Chest Hospital, 215 Guangzhou Road, Nanjing, 210029, China
| | | | | | | | | |
Collapse
|
22
|
Masago K, Fujimoto D, Fujita S, Hata A, Kaji R, Ohtsuka K, Okuda C, Takeshita J, Katakami N. Response to bevacizumab combination chemotherapy of malignant pleural effusions associated with non-squamous non-small-cell lung cancer. Mol Clin Oncol 2014; 3:415-419. [PMID: 25798278 DOI: 10.3892/mco.2014.457] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 10/29/2014] [Indexed: 02/01/2023] Open
Abstract
Malignant pleural effusion (MPE) is a common complication of lung cancer with devastating consequences. Since vascular endothelial growth factor (VEGF) has been implicated in MPE, we hypothesized that bevacizumab, an anti-VEGF antibody, may be effective against MPE in patients with non-small-cell lung cancer (NSCLC). We analysed the records of 21 patients treated for NSCLC-associated MPE between February, 2010 and August, 2013 who consequently underwent bevacizumab combination chemotherapy at the Institute of Biomedical Research and Innovation Hospital. The results were retrospectively analysed using case records and radiographic imaging records. Three patients exhibited complete response of the pleural effusion to bevacizumab treatment, 8 patients achieved a partial response (PR) and 6 patients showed no response. When efficacy was assessed by the response of the measurable primary or metastatic lesions to the treatment, 5 patients achieved a PR, 13 patients had stable disease and 3 patients exhibited progressive disease. The response rate (RR) of the pleural effusion to the antibody treatment was 71.4% and the overall RR of measurable lesions was 23.8%. The median time-to-response for pleural effusion was 132 days. In conclusion, this study demonstrated a high R R to bevacizumab combination therapy for the MPE associated with non-squamous NSCLC. Therefore, bevacizumab therapy may be considered a therapeutic option for patients with non-squamous NSCLC who develop MPE.
Collapse
Affiliation(s)
- Katsuhiro Masago
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047 ; Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Aichi 464-8681
| | - Daichi Fujimoto
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Hyogo 650-0047
| | - Shiro Fujita
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| | - Akito Hata
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| | - Reiko Kaji
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| | - Kyoko Ohtsuka
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047 ; Department of Respiratory Medicine, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Chiyuki Okuda
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| | - Jumpei Takeshita
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| | - Nobuyuki Katakami
- Division of Integrated Oncology, Institute of Biomedical Research and Innovation Hospital, Kobe, Hyogo 650-0047
| |
Collapse
|
23
|
Lv M, Xu Y, Tang R, Ren J, Shen S, Chen Y, Liu B, Hou Y, Wang T. miR141–CXCL1–CXCR2 Signaling–Induced Treg Recruitment Regulates Metastases and Survival of Non–Small Cell Lung Cancer. Mol Cancer Ther 2014; 13:3152-62. [DOI: 10.1158/1535-7163.mct-14-0448] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Xu C, Yu L, Zhan P, Zhang Y. Elevated pleural effusion IL-17 is a diagnostic marker and outcome predictor in lung cancer patients. Eur J Med Res 2014; 19:23. [PMID: 24887477 PMCID: PMC4041345 DOI: 10.1186/2047-783x-19-23] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/02/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Interleukin 17 (IL-17) is a proinflammatory cytokine produced mainly by CD4(+) T-lymphocytes and may be important in tumor cell growth and progression. In this study, we aimed to evaluate the diagnostic and prognostic value of pleural effusion levels of IL-17 in lung cancer patients with malignant pleural effusion (MPE). METHODS Pleural effusion samples were collected from 78 lung cancer patients with MPE and from 45 patients with nonmalignant pleural effusion. Pleural fluid concentrations of IL-17 were measured by using enzyme-linked immunosorbent assays. RESULTS Malignant effusion exhibited higher IL-17 levels than nonmalignant effusion (20.49 ± 5.27 pg/ml vs. 13.16 ± 2.25 pg/ml; P < 0.01). Lung cancer patients with pleural fluid IL-17 levels below 15 pg/ml had longer overall survival than those patients with higher levels (10.8 months vs. 4.7 months; P < 0.05). On the basis of multivariate analysis, we found that pleural fluid IL-17 level was an independent prognostic factor in lung cancer patients with MPE. CONCLUSIONS Measurement of IL-17 levels might be a useful diagnostic and prognostic test for lung cancer patients with MPE.
Collapse
Affiliation(s)
| | - LiKe Yu
- First Department of Respiratory Medicine, Nanjing Chest Hospital, 215 Guangzhou Road, Nanjing 210029, China.
| | | | | |
Collapse
|
25
|
Yeh HH, Chang WT, Lu KC, Lai WW, Liu HS, Su WC. Upregulation of tissue factor by activated Stat3 contributes to malignant pleural effusion generation via enhancing tumor metastasis and vascular permeability in lung adenocarcinoma. PLoS One 2013; 8:e75287. [PMID: 24086497 PMCID: PMC3785526 DOI: 10.1371/journal.pone.0075287] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/13/2013] [Indexed: 11/19/2022] Open
Abstract
Malignant pleural effusion (MPE) is a poor prognostic sign for patients with lung cancer. Tissue factor (TF) is a coagulation factor that participates in angiogenesis and vascular permeability and is abundant in MPE. We previously demonstrated that autocrine IL-6-activated Stat3 contributes to tumor metastasis and upregulation of VEGF, resulting in the generation of MPE in lung adenocarcinoma. In this study, we found IL-6-triggered Stat3 activation also induces TF expression. By using pharmacologic inhibitors, it was shown that JAK2 kinase, but not Src kinase, contributed to autocrine IL-6-induced TF expression. Inhibition of Stat3 activation by dominant negative Stat3 (S3D) in lung adenocarcinoma suppressed TF-induced coagulation, anchorage-independent growth in vitro, and tumor growth in vivo. Consistently, knockdown of TF expression by siRNA resulted in a reduction of anchorage-independent growth of lung adenocarcinoma cells. Inhibition of TF expression also decreased the adhesion ability of cancer cells in normal lung tissues. In the nude mouse model, both lung metastasis and MPE generation were decreased when PC14PE6/AS2-siTF cells (TF expression was silenced) were intravenously injected. PC14PE6/AS2-siTF cells also produced less malignant ascites through inhibition of vascular permeability. In summary, we showed that TF expression plays a pivotal role in the pathogenesis of MPE generation via regulating of tumor metastasis and vascular permeability in lung adenocarcinoma bearing activated Stat3.
Collapse
Affiliation(s)
- Hsuan-Heng Yeh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cancer Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tsan Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuang-Chu Lu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Wei Lai
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Sheng Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center for Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (WCS); (HSL)
| | - Wu-Chou Su
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Cancer Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (WCS); (HSL)
| |
Collapse
|
26
|
Chen SF, Lin YS, Jao SW, Chang YC, Liu CL, Lin YJ, Nieh S. Pulmonary Adenocarcinoma in Malignant Pleural Effusion Enriches Cancer Stem Cell Properties during Metastatic Cascade. PLoS One 2013; 8:e54659. [PMID: 23658677 PMCID: PMC3641054 DOI: 10.1371/journal.pone.0054659] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 12/17/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metastasis occurs in a series of discrete steps involving invasion, angiogenesis, lymphovascular space permeation, and establishment of secondary tumors. Malignant pleural effusion (MPE), a type of tumor metastasis, is usually a poor prognostic sign for patients with pulmonary adenocarcinoma, although its underlying mechanism has received less attention than other types of metastases have. The objective of the current study was to confirm whether cancer stem cells (CSCs) in MPE contribute to the "metastatic cascade" through the epithelial - mesenchymal transition (EMT), anoikis, and adaptation in the microenvironment. METHODS Pulmonary tissue and corresponding cell blocks of MPE samples from 20 patients with primary adenocarcinoma were analyzed by immunohistochemical staining with CSC-representative markers (CD133, Nanog, and OCT-4) and EMT-associated markers (E-cadherin and vimentin). Correlations between these variables and clinico-pathological parameters were analyzed. Primary cultures from eight cases of MPE were investigated to characterize the CSC properties, including marker expression, sphere formation, and differentiation. RESULTS Expressions of CSC-representative markers for 20 cases of MPE cell blocks were quite diverse and variable ranging from 15% to 90%. Stronger expression of CSC-representative markers and alteration of EMT-associated markers were found at the invasive fronts and in MPEs compared with the expression in primary pulmonary tumor tissues. The expression of OCT-4 in MPEs significantly related to distant metastasis and stage, as well as inversely correlated with patient survival. Primary cultures confirmed the CSC properties in MPE. Five of eight cases of MPE yielded adequate cell clusters, which also showed variable expressions of CSC markers in addition to sphere formation and the ability for differentiation and metastasis. CONCLUSION This pilot study offers a better understanding of the metastatic cascade. Establishing a model of MPE will provide further insight into the role of CSCs in metastasis and may explain the high therapeutic failure rates for patients with MPE.
Collapse
Affiliation(s)
- Su-Feng Chen
- Department of Dental Hygiene, China Medical University, Taichung, Taiwan
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yaoh-Shiang Lin
- Department of Otolaryngology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shu-Wen Jao
- Division of Colon and Rectal Surgery, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yun-Ching Chang
- Graduate Institute of Life Sciences, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Chia-Lin Liu
- Graduate Institute of Life Sciences, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yu-Ju Lin
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Shin Nieh
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
27
|
Interleukin-8 (CXCL8) in tumor associated non-vascular extracellular fluids: its diagnostic and prognostic values. A review. Int J Biol Markers 2012; 27:169-78. [PMID: 22610755 DOI: 10.5301/jbm.2012.9261] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2012] [Indexed: 12/25/2022]
Abstract
Interleukin-8 (IL-8, CXCL8) was originally discovered as a powerful attractor and activator of polymorphonuclear leukocytes. It was soon recognized that IL-8 also affects proliferation and migration of cancer cells, tumor angiogenesis and metastasis, and that it is expressed in many cancerous cell types. IL-8 protein expression is usually increased in serum of cancer patients, but markedly higher concentrations are found in cancer-associated non-vascular extracellular fluids, such as pleural effusion, ascites and cyst fluid. Elevated concentrations of IL-8 are indicative of malignant processes also in cerebrospinal fluid, urine, saliva, interstitial fluid and cervicovaginal secretions. Higher IL-8 levels are typically found in high-grade peritumoral fluids rather than low-grade tumors and benign conditions, with the exception of inflammatory processes. In line with recent molecular biology investigations, it appears that the local IL-8 production is related to its malignant origin and to tumor progression. Hence, IL-8 in peritumoral fluid is to be taken into consideration while assessing tumor character and monitoring the tumor progression/remission status. Besides, the data here collected justify the attempts to find an IL-8-targeted inhibitory therapy
Collapse
|
28
|
Stathopoulos GT, Kalomenidis I. Malignant pleural effusion: tumor-host interactions unleashed. Am J Respir Crit Care Med 2012; 186:487-92. [PMID: 22652027 PMCID: PMC5650050 DOI: 10.1164/rccm.201203-0465pp] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Malignant pleural effusion (MPE) poses a significant clinical problem. Current nonetiologic management is suboptimal in terms of efficacy and safety. In light of recent research progress, we propose herein a new view of MPE development, which may rapidly translate into meaningful changes in therapeutics. In addition to tumor-induced impairment of pleural fluid drainage, pertinent findings point toward another pathway to MPE formation: a vicious loop of interactions between pleural-based tumor cells and the host vasculature and immune system that results in increased net fluid production via enhanced plasma extravasation into the pleural space. The ability of tumor cells to trigger this cascade likely rests on a specific and distinct transcriptional repertoire, which results in important vasoactive events in the pleural space. Although the characterization of tumor-derived factors responsible for MPE development is in the making, an additional, indirect path to MPE was recently demonstrated: tumor cells recruit and co-opt host cells and mediators, which, in turn, amplify tumor cell-primed fluid leakage and impact tumor cell functions. Importantly, recent evidence suggests that the biologic events that culminate in clinical MPE are likely amenable to therapeutic inhibition and even prevention. In this perspective, the scientific basis for an update of current concepts of MPE formation is highlighted. Key questions for future research are posed. Finally, a vision for novel, effective, safe, and convenient treatment modalities that can be offered to outpatients with MPE is set forth.
Collapse
Affiliation(s)
- Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Basic Biomedical Sciences Building, 2nd floor, Room B40, 1 Asklepiou Street, University Campus (Panepistimioupolis), 26504 Rio, Greece.
| | | |
Collapse
|
29
|
Cell-free microRNA expression profiles in malignant effusion associated with patient survival in non-small cell lung cancer. PLoS One 2012; 7:e43268. [PMID: 22937028 PMCID: PMC3427341 DOI: 10.1371/journal.pone.0043268] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/18/2012] [Indexed: 01/14/2023] Open
Abstract
Objective MicroRNAs (miRNAs) expression is altered in cancer cells, and miRNAs could serve as diagnostic and prognostic biomarker for cancer patients. This study was designed to analyze circulating miRNAs expression in the malignant pleural effusion (MPE) and their association with patient survival in non-small cell lung cancer (NSCLC). Methods Pleural effusion from 184 patients with NSCLC and MPE were collected. MiRNA microarray and bioinformatics interpretation were used to evaluate miRNA expression profiles in 10 NSCLC patients with different survival prognosis. Associations were validated in 184 patients (randomly classified into training and validation set with equal number in each group) using quantitative RT-PCR. Risk scores were formulated based on the expression signature of miRNAs. Clinical data, such as patient survival, were collected for correlation analysis. Results Thirty-three miRNAs were found to be altered more than two-fold by microarray in malignant effusions between longer-survival and shorter-survival groups, and levels of five miRNAs (miRNA-93, miRNA-100, miRNA-134, miRNA-151 and miRNA-345) were significantly associated with overall survival. High expression of miR-100 and low expression of miRNA-93, miRNA-134, miRNA-151 and miRNA-345 were associated with poor survival in both the training and validation cohort. Patients with high risk scores had overall poor survival compared to the patients with low risk scores. Risk score was an independent predictor of patient survival. Conclusions Expression patterns of miRNAs are systematically altered in MPE of patient with NSCLC. The five miRNA signature from the effusion may serve as a predictor for the overall survival of patients with lung cancers.
Collapse
|
30
|
Froudarakis ME. Pleural Effusion in Lung Cancer: More Questions than Answers. Respiration 2012; 83:367-76. [DOI: 10.1159/000338169] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
31
|
Salgia R. Prognostic significance of angiogenesis and angiogenic growth factors in nonsmall cell lung cancer. Cancer 2011; 117:3889-99. [PMID: 21858799 PMCID: PMC3160199 DOI: 10.1002/cncr.25935] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 11/30/2010] [Accepted: 12/16/2010] [Indexed: 01/22/2023]
Abstract
Currently, nonsmall-cell lung cancer (NSCLC) is the leading cause of cancer-related death in the United States. Angiogenesis, the formation of new vasculature, is a complex and tightly regulated process that promotes metastasis and disease progression in lung cancer and other malignancies. Developmental antiangiogenic agents have shown activity in NSCLC, and bevacizumab, an antiangiogenic monoclonal antibody, is approved for the treatment of patients with advanced disease. However, predictive biomarkers are needed to guide the administration of antiangiogenic agents. It is possible that angiogenic molecules could accurately predict patient response to targeted antiangiogenic therapies, which would allow individualized and perhaps more effective treatment. Angiogenic signaling molecules may also have value as prognostic indicators, which may be useful for the management of NSCLC. Here the author provides an overview of angiogenic molecules currently being investigated as prognostic biomarkers in NSCLC and discusses their potential to guide treatment choices.
Collapse
Affiliation(s)
- Ravi Salgia
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
32
|
See KC, Lee P. Advances in the diagnosis of pleural disease in lung cancer. Ther Adv Respir Dis 2011; 5:409-18. [DOI: 10.1177/1753465811408637] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pleural disease in lung cancer can be benign or malignant with the latter carrying a grave prognosis. In this review, we describe and discuss the advances in pleural imaging, procedures, and biomarkers for the diagnosis of pleural diseases in lung cancer. Ultrasound and computed tomography are increasingly applied in the planning of pleural procedures to enhance diagnostic accuracy and safety whilst pleuroscopy gives excellent yield in excess of 93% in the evaluation of cytology negative pleural effusions. Invasion beyond the elastic layer of the visceral pleura upstages lung cancer, and may indicate a need for adjuvant chemotherapy. Biomarkers isolated from pleural fluid or tissue may aid in diagnosis and guide treatment in the future. Magnetic resonance imaging, positron emission tomography, narrow band imaging of the pleura and autofluorescence thoracoscopy are technologies that require further evaluation to better define their respective roles in the diagnostic algorithms of pleural diseases in lung cancer.
Collapse
|
33
|
Sakr L, Maldonado F, Greillier L, Dutau H, Loundou A, Astoul P. Thoracoscopic assessment of pleural tumor burden in patients with malignant pleural effusion: prognostic and therapeutic implications. J Thorac Oncol 2011; 6:592-7. [PMID: 21258256 DOI: 10.1097/jto.0b013e318208c7c1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Malignant pleural effusion (MPE) is encountered at an advanced stage of disease progression and often heralds a poor prognosis. The most reliable predictive factor of survival in such patients is the primary tumor. Thoracoscopy is often performed for accurate diagnosis and/or thoracoscopic talc insufflation as a therapeutic modality. It remains unknown whether pleural tumor burden, as visualized on thoracoscopy, has potential prognostic value. The objective of this study was to determine the prognostic accuracy of pleural tumor extent and localization (parietal, visceral, or diaphragmatic involvement), as assessed during medical thoracoscopy. METHODS Medical records of all patients who underwent thoracoscopy for suspicion of MPE between 2001 and 2008 at a tertiary care referral hospital were reviewed. Patients were included if pleural metastatic invasion was confirmed on tissue biopsy and survival status ascertained. RESULTS Four hundred twenty-one patients underwent diagnostic or therapeutic medical thoracoscopy at our referral center. Among them, 122 had confirmed metastatic pleural spread, but survival data were lacking in 15. Primary tumor consisted of non-mall cell lung cancer in 56, breast cancer in 23, melanoma in eight, and other malignancies in 20. Median survival of the entire population was 9.4 months. On univariate analysis, the following variables were significantly associated with reduced median overall survival: pleural metastatic melanoma, age less than 60 years, bloody MPE, extensive pleural adhesions, and widespread visceral pleural nodules (p < 0.05). On multivariate analysis, only melanoma as a primary tumor, pleural fluid appearance and extent of pleural adhesions remained independent and significant predictors of survival. CONCLUSION No significant association was found between the extent or localization of pleural tumor burden and overall survival.
Collapse
Affiliation(s)
- Lama Sakr
- Division of Pulmonary Diseases, Sir Mortimer B. Davis Jewish General Hospital, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Sriram KB, Relan V, Clarke BE, Duhig EE, Yang IA, Bowman RV, Lee YCG, Fong KM. Diagnostic molecular biomarkers for malignant pleural effusions. Future Oncol 2011; 7:737-52. [DOI: 10.2217/fon.11.45] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Malignant pleural effusions (MPEs) are a common and important cause of cancer-related mortality and morbidity. Prompt diagnosis using minimally invasive tests is important because the median survival after diagnosis is only 4–9 months. Pleural fluid cytology is pivotal to current MPE diagnostic algorithms but has limited sensitivity (30–60%). Consequently, many patients need to undergo invasive diagnostic tests such as thoracoscopic pleural biopsy. Recent genomic, transcriptomic, methylation and proteomic studies on cells within pleural effusions have identified novel molecular diagnostic biomarkers that demonstrate potential in complementing cytology in the diagnosis of MPEs. Several challenges will need to be addressed prior to the incorporation of these molecular tests into routine clinical diagnosis, including validation of molecular diagnostic markers in well-designed prospective, comparative and cost–effectiveness studies. Ultimately, minimally invasive diagnostic tests that can be performed quickly will enable clinicians to provide the most effective therapies for patients with MPEs in a timely fashion.
Collapse
Affiliation(s)
| | - Vandana Relan
- University of Queensland Thoracic Research Centre, School of Medicine, The University of Queensland, Queensland, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Queensland, Australia
| | - Belinda E Clarke
- Department of Anatomical Pathology, The Prince Charles Hospital, Queensland, Australia
| | - Edwina E Duhig
- Department of Anatomical Pathology, The Prince Charles Hospital, Queensland, Australia
| | - Ian A Yang
- University of Queensland Thoracic Research Centre, School of Medicine, The University of Queensland, Queensland, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Queensland, Australia
| | - Rayleen V Bowman
- University of Queensland Thoracic Research Centre, School of Medicine, The University of Queensland, Queensland, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Queensland, Australia
| | - YC Gary Lee
- School of Medicine & Pharmacology & CAARR, University of Western Australia, Perth, Australia
- Respiratory Department, Sir Charles Gairdner Hospital, Perth, Australia
- Pleural Disease Unit, Lung Institute of Western Australia, Perth, Australia
| | - Kwun M Fong
- University of Queensland Thoracic Research Centre, School of Medicine, The University of Queensland, Queensland, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Queensland, Australia
| |
Collapse
|
35
|
Chen Y, Liang B, Zhao YJ, Wang SC, Fan YB, Wu GP. Transcription expression and clinical significance of vascular endothelial growth factor mRNA and endostatin mRNA in pleural effusions of patients with lung cancer. Diagn Cytopathol 2010; 40:287-91. [PMID: 22431315 DOI: 10.1002/dc.21546] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 08/19/2010] [Indexed: 11/07/2022]
Abstract
The aim of this study was to evaluate the individual and combined diagnostic utility of vascular endothelial growth factor (VEGF) mRNA and endostatin mRNA in pleural effusions of patients with lung cancer. Transcription levels of VEGF and endostatin were detected by reverse transcription polymerase chain reaction (RT-PCR) in pleural effusions of patients with lung cancer (92 cases) and with lung benign disease (36 cases). Both VEGF mRNA and endostatin mRNA was significantly higher in malignant, AC, and SCC effusions than in benign effusions (P < 0.01). In the subgrouping, VEGF mRNA was obviously higher than endostatin mRNA in malignant and AC effusions (P < 0.01), whereas VEGF mRNA and endostatin mRNA did not differ between AC group and SCC group (P > 0.05). In single, VEGF mRNA had the highest sensitivity (82.6%) and accuracy (84.3%), whereas endostatin mRNA had the highest specificity (100%). When combinations of VEGF mRNA and endostatin mRNA were evaluated together, they gave a high-diagnostic performance: sensitivity of 95.7% and accuracy of 93.8%, respectively. The detection of VEGF mRNA and endostatin mRNA appears to be suitable for distinguishing carcinoma cells from reactive mesothelial cells in pleural effusions, they could be useful to diagnose the pleural micrometastasis.
Collapse
Affiliation(s)
- Ying Chen
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
36
|
CD105 (Endoglin) expression in breast carcinoma effusions is a marker of poor survival. Breast 2010; 19:493-8. [PMID: 21078485 DOI: 10.1016/j.breast.2010.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 05/24/2010] [Indexed: 12/19/2022] Open
Abstract
We analyzed the expression and clinical role of endoglin (CD105) in breast carcinoma effusions. Endoglin levels were measured in 36 effusion supernatants by ELISA and studied for association with the cancer-associated markers calprotectin, VEGF, and the VEGF receptor sFlt1. Endoglin expression was further studied in 46 effusions and 22 primary carcinomas using immunohistochemistry. The four secreted molecules were detected in all specimens and their levels significantly correlated (p < 0.001). In effusions, endoglin was localized to carcinoma cells and reactive mesothelium using immunohistochemistry. Tumor cell expression was higher in effusions compared to primary carcinomas (p = 0.025), and in post-chemotherapy compared to pre-chemotherapy effusions (p = 0.017). Higher tumor endoglin expression was associated with poor overall (p = 0.021) and disease-free (p = 0.032) survival in univariate analysis, and was an independent predictor in Cox multivariate analysis (p = 0.001 and p = 0.038, respectively). Our data suggest that endoglin may be an important therapeutic target in metastatic breast cancer.
Collapse
|