51
|
Rodríguez-Berriguete G, Sánchez-Espiridión B, Cansino JR, Olmedilla G, Martínez-Onsurbe P, Sánchez-Chapado M, Paniagua R, Fraile B, Royuela M. Clinical significance of both tumor and stromal expression of components of the IL-1 and TNF-α signaling pathways in prostate cancer. Cytokine 2013; 64:555-63. [DOI: 10.1016/j.cyto.2013.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/05/2013] [Accepted: 09/01/2013] [Indexed: 10/26/2022]
|
52
|
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
|
53
|
Marazioti A, Kairi CA, Spella M, Giannou AD, Magkouta S, Giopanou I, Papaleonidopoulos V, Kalomenidis I, Snyder LA, Kardamakis D, Stathopoulos GT. Beneficial impact of CCL2 and CCL12 neutralization on experimental malignant pleural effusion. PLoS One 2013; 8:e71207. [PMID: 23967166 PMCID: PMC3743892 DOI: 10.1371/journal.pone.0071207] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022] Open
Abstract
Using genetic interventions, we previously determined that C-C motif chemokine ligand 2 (CCL2) promotes malignant pleural effusion (MPE) formation in mice. Here we conducted preclinical studies aimed at assessing the specific therapeutic potential of antibody-mediated CCL2 blockade against MPE. For this, murine MPEs or skin tumors were generated in C57BL/6 mice by intrapleural or subcutaneous delivery of lung (LLC) or colon (MC38) adenocarcinoma cells. Human lung adenocarcinoma cells (A549) were used to induce MPEs in severe combined immunodeficient mice. Intraperitoneal antibodies neutralizing mouse CCL2 and/or CCL12, a murine CCL2 ortholog, were administered at 10 or 50 mg/kg every three days. We found that high doses of CCL2/12 neutralizing antibody treatment (50 mg/kg) were required to limit MPE formation by LLC cells. CCL2 and CCL12 blockade were equally potent inhibitors of MPE development by LLC cells. Combined CCL2 and CCL12 neutralization was also effective against MC38-induced MPE and prolonged the survival of mice in both syngeneic models. Mouse-specific CCL2-blockade limited A549-caused xenogeneic MPE, indicating that host-derived CCL2 also contributes to MPE precipitation in mice. The impact of CCL2/12 antagonism was associated with inhibition of immune and vascular MPE-related phenomena, such as inflammation, new blood vessel assembly and plasma extravasation into the pleural space. We conclude that CCL2 and CCL12 blockade are effective against experimental MPE induced by murine and human adenocarcinoma in mice. These results suggest that CCL2-targeted therapies may hold promise for future use against human MPE.
Collapse
Affiliation(s)
- Antonia Marazioti
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Chrysoula A. Kairi
- First Department of Critical Care and Pulmonary Medicine, University of Athens School of Medicine, General Hospital Evangelismos, Athens, Attica, Greece
| | - Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Anastasios D. Giannou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Sophia Magkouta
- First Department of Critical Care and Pulmonary Medicine, University of Athens School of Medicine, General Hospital Evangelismos, Athens, Attica, Greece
| | - Ioanna Giopanou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Vassilios Papaleonidopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Ioannis Kalomenidis
- First Department of Critical Care and Pulmonary Medicine, University of Athens School of Medicine, General Hospital Evangelismos, Athens, Attica, Greece
| | - Linda A. Snyder
- Janssen R&D, LLC, Oncology Discovery Research, Spring House, Pennsylvania, United States of America
| | - Dimitrios Kardamakis
- Department of Radiation Oncology and Stereotactic Radiotherapy, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
| | - Georgios T. Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, Achaia, Greece
- First Department of Critical Care and Pulmonary Medicine, University of Athens School of Medicine, General Hospital Evangelismos, Athens, Attica, Greece
- * E-mail:
| |
Collapse
|
54
|
Ma X, Sun Y, Wang S, Yang Z, Song Y. [Establishment of a malignant pleural effusion mouse model with Lewis lung carcinoma cell lines expressing enhanced green fluorescent protein]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 15:317-23. [PMID: 22681917 PMCID: PMC6000302 DOI: 10.3779/j.issn.1009-3419.2012.06.01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
背景与目的 恶性胸腔积液是晚期肺癌预后差的因素之一。本研究拟用Lewis肺癌细胞构建裸鼠恶性胸腔积液模型,从而建立一个良好的动物实验研究平台。 方法 经胸膜腔接种表达增强型绿色荧光蛋白的Lewis肺癌细胞,建立裸鼠恶性胸腔积液模型。定期解剖小鼠,经小动物活体荧光成像系统观察肿瘤的生长。剩余小鼠定期行胸部CT检查,计算各时间点的成胸水率,并观察建模后裸鼠的生存期和肿瘤转移情况。所有小鼠在解剖时发现有胸水,抽取并计量,同一时间点内获得多份胸水标本,计算其平均体积。利用相关性检验分析胸水体积与肿瘤积分光密度之间的相关性。 结果 接种后第4天,荧光体视镜下可发现胸膜上有绿色荧光,成瘤率100%。随接种时间延长,肿瘤体积逐渐增加,肿瘤侵及纵隔和肺门淋巴结、对侧胸膜、心包,转移率分别为87%、73%和20%。第7天、第14天和第21天成胸水率分别为13%、46%和53%。小鼠平均生存时间为28.8天,所有胸水均为血性,胸水平均体积在第10天以后逐渐增加,第16天达到峰值(0.5 mL)。胸水体积与积分光密度之间具有相关性(r=0.91, P < 0.000, 1)。 结论 本研究将表达增强型绿色荧光蛋白的肺癌细胞在显微镜下经胸膜腔接种成功建立肺癌恶性胸腔积液模型,有助于动态观察肿瘤细胞在胸腔内的生物学行为,该模型可应用于肺癌的基础研究及抗肿瘤药物开发。
Collapse
Affiliation(s)
- Xingqun Ma
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Clinical School of the Medical College of
Nanjing University, Nanjing 210002, China
| | | | | | | | | |
Collapse
|
55
|
Abstract
Active, but dysfunctional, immune responses in patients with cancer have been studied in several tumour types, but owing to the heterogeneity of cancer theories of common reaction mechanisms seem to be obsolete. In this Review of published clinical studies of patients with cancer, expression and interplay of the following cytokines are examined: interleukin 2, interleukin 6, interleukin 8, interleukin 10, interleukin 12, interleukin 18, tumour necrosis factor α (TNFα), transforming growth factor β (TGFβ), interferon-γ, HLA-DR, macrophage migration inhibitory factor (MIF), and C-X-C motif chemokine receptor 4 (CXCR4). Clinical data were analysed in a non-quantitative descriptive manner and interpreted with regard to experimentally established physiological cytokine interactions. The clinical cytokine pattern that emerged suggests that simultaneous immunostimulation and immunosuppression occur in patients with cancer, with increased concentrations of the cytokines MIF, TNFα, interleukin 6, interleukin 8, interleukin 10, interleukin 18, and TGFβ. This specific cytokine pattern seems to have a prognostic effect, since high interleukin 6 or interleukin 10 serum concentrations are associated with negative prognoses in independent cancer types. Although immunostimulatory cytokines are involved in local cancer-associated inflammation, cancer cells seem to be protected from immunological eradication by cytokine-mediated local immunosuppression and a resulting defect of the interleukin 12-interferon-γ-HLA-DR axis. Cytokines produced by tumours might have a pivotal role in this defect. A working hypothesis is that the cancer-specific and histology-independent uniform cytokine cascade is one of the manifestations of the underlying paraneoplastic systemic disease, and this hypothesis links the stage of cancer with both the functional status of the immune system and the patient's prognosis. Neutralisation of this cytokine pattern could offer novel and so far unexploited treatment approaches for cancer.
Collapse
|
56
|
Bradshaw M, Mansfield A, Peikert T. The role of vascular endothelial growth factor in the pathogenesis, diagnosis and treatment of malignant pleural effusion. Curr Oncol Rep 2013; 15:207-16. [PMID: 23568600 PMCID: PMC3674487 DOI: 10.1007/s11912-013-0315-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Malignant pleural effusions (MPEs) are a significant source of cancer-related morbidity. Over 150,000 patients in the United States suffer from breathlessness and diminished quality of life due to MPE each year. Current management strategies are of mostly palliative value and focus on symptom control; they do not address the pathobiology of the effusion, nor do they improve survival. Further elucidation of the pathophysiological mechanisms, coupled with the development of novel treatments such as intrapleural chemotherapeutics targeting this process, has the potential to greatly improve the efficacy of our current management options. Vascular endothelial growth factor-A (VEGF-A) has been implicated as a critical cytokine in the formation of malignant pleural effusions. Elevated levels of VEGF produced by tumor cells, mesothelial cells, and infiltrating immune cells result in increased vascular permeability, cancer cell transmigration, and angiogenesis. Therefore antiangiogenic therapies such as Bevacizumab, a monoclonal antibody targeting VEGF-A, may have a potential role in the management of malignant pleural effusions. Herein we review the pathogenesis and potential treatment strategies of malignant pleural effusions, with a focus on angiogenesis and antiangiogenic therapeutics.
Collapse
|
57
|
Lee CH, Lin SH, Chang SF, Chang PY, Yang ZP, Lu SC. Extracellular signal-regulated kinase 2 mediates the expression of granulocyte colony-stimulating factor in invasive cancer cells. Oncol Rep 2013; 30:419-24. [PMID: 23674093 DOI: 10.3892/or.2013.2463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/19/2013] [Indexed: 11/06/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) affects granulopoiesis and is important for mobilizing neutrophils into blood circulation. Due to the hematopoietic properties of G-CSF, it has been widely used to clinically treat chemotherapy-induced neutropenia. However, G-CSF can promote tumors by inhibiting innate and adaptive immunity and enhancing angiogenesis and neoplastic growth. Most G-CSF-producing tumors are associated with a poor prognosis. This indicates that G-CSF promotes cancer progression. Thus, identifying regulatory molecules involved in tumor-derived G-CSF expression may provide therapeutic targets for cancer treatment. This study identified considerable G-CSF expression in malignant breast, lung and oral cancer cells. However, G-CSF expression was barely detectable in non-invasive cell lines. Expression of G-CSF mRNA and protein increased during exposure to tumor necrosis factor-α (TNF-α). Treatment with U0126 (a mitogen-activated protein kinase inhibitor) drastically reduced basal levels of G-CSF and TNF-α-induced G-CSF in aggressive cancer cells. This study also showed that knockdown of extracellular signal-regulated kinase (ERK) 2 by shRNA was necessary and sufficient to eliminate the expression of tumor-derived G-CSF. This did not apply to ERK1. Therefore, ERK2 (but not ERK1) is responsible for the transcriptional regulation of tumor-derived G-CSF. The results indicate the pharmaceutical value of specific ERK2 inhibitors in treating patients with G-CSF-producing tumors.
Collapse
Affiliation(s)
- Chia-Huei Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan, R.O.C
| | | | | | | | | | | |
Collapse
|
58
|
Ma X, Yao Y, Yuan D, Liu H, Wang S, Zhou C, Song Y. Recombinant human endostatin endostar suppresses angiogenesis and lymphangiogenesis of malignant pleural effusion in mice. PLoS One 2012; 7:e53449. [PMID: 23285296 PMCID: PMC3532165 DOI: 10.1371/journal.pone.0053449] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 11/28/2012] [Indexed: 01/02/2023] Open
Abstract
Background Malignant pleural effusion (MPE) is a common complication of lung cancer. One widely used treatment for MPE is Endostar, a recombined humanized endostatin based treatment. However, the mechanism of this treatment is still unclear. The aim of this study was to investigate the effects of Endostar in mice with MPE. Methods and Materials Lewis lung carcinoma (LLC) cell line expressing enhanced green fluorescent protein (EGFP) was injected into pleural cavity to establish MPE mice model. Mice were randomly divided into four groups. High dose of Endostar (30 mg/kg), low dose of Endostar (8 mg/kg), normal saline, or Bevacizumab (5 mg/kg) was respectively injected into pleural cavity three times with 3-day interval in each group. Transverse computed tomography (CT) was performed to observe pleural fluid formation 14 days after LLC cells injection. Mice were anesthetized and sacrificed 3 days after final administration. The volume of pleural effusion n was measured using 1 ml syringe. Micro blood vessel density (MVD), Lymphatic micro vessel density (LMVD), the expression level of vascular endothelial growth factor A (VEGF-A) and VEGF-C were observed by immunohistochemistry (IHC) staining. Results The volume of pleural effusion as well as the number of pleural tumor foci, MVD and the expression of VEGF-A were significantly reduced in high dose of Endostar treat group. More importantly, LMVD and the expression of VEGF-C were markedly lower in treat group than those in the other three control groups. Conclusion Our work demonstrated that Endostar played an efficient anti-cancer role in MPE through its suppressive effect on angiogenesis and lymphangiogenesis, which provided a certain theoretical basis for the effectiveness of Endostar on the MPE treatment.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/complications
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Down-Regulation/drug effects
- Drug Evaluation, Preclinical
- Endostatins/pharmacology
- Endostatins/therapeutic use
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Lymphangiogenesis/drug effects
- Lymphangiogenesis/physiology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Transplantation
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/prevention & control
- Pleural Effusion, Malignant/complications
- Pleural Effusion, Malignant/drug therapy
- Pleural Effusion, Malignant/pathology
- Recombinant Proteins/pharmacology
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Xingqun Ma
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yanwen Yao
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Shouju Wang
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- * E-mail:
| |
Collapse
|
59
|
Abstract
Solid tumors consist of neoplastic cells, non-malignant stromal cells, and migratory hematopoietic cells. Complex interactions between the cell types in this microenvironment regulate tumor growth, progression, metastasis, and angiogenesis. The cells and mediators of inflammation form a major part of the epithelial tumor microenvironment. In some cancers, inflammatory conditions precede development of malignancy; in others, oncogenic change drives a tumor-promoting inflammatory milieu. Whatever its origin, this "smoldering" inflammation aids proliferation and survival of malignant cells, stimulates angiogenesis and metastasis, subverts adaptive immunity, and alters response to hormones and chemotherapy. Cytokines are major mediators of communication between cells in the inflammatory tumor microenvironment. It is known that neoplastic cells often over-express proinflammatory mediators including proteases, eicosanoids, cytokines, and chemokines. Several cytokines such as macrophage migratory inhibitory factor (MIF), TNF-α, IL-6, IL-17, IL-12, IL-23, IL-10, and TGF-β have been linked with both experimental and human cancers and can either promote or inhibit tumor development. MIF is a major cytokine in many cancers and there is evidence that the cytokine is produced by both malignant cells and infiltrating leukocytes. In this article we will discuss the role of cancer-associated inflammation and the particular role of MIF in malignant disease.
Collapse
|
60
|
Abstract
Solid tumors consist of neoplastic cells, non-malignant stromal cells, and migratory hematopoietic cells. Complex interactions between the cell types in this microenvironment regulate tumor growth, progression, metastasis, and angiogenesis. The cells and mediators of inflammation form a major part of the epithelial tumor microenvironment. In some cancers, inflammatory conditions precede development of malignancy; in others, oncogenic change drives a tumor-promoting inflammatory milieu. Whatever its origin, this "smoldering" inflammation aids proliferation and survival of malignant cells, stimulates angiogenesis and metastasis, subverts adaptive immunity, and alters response to hormones and chemotherapy. Cytokines are major mediators of communication between cells in the inflammatory tumor microenvironment. It is known that neoplastic cells often over-express proinflammatory mediators including proteases, eicosanoids, cytokines, and chemokines. Several cytokines such as macrophage migratory inhibitory factor (MIF), TNF-α, IL-6, IL-17, IL-12, IL-23, IL-10, and TGF-β have been linked with both experimental and human cancers and can either promote or inhibit tumor development. MIF is a major cytokine in many cancers and there is evidence that the cytokine is produced by both malignant cells and infiltrating leukocytes. In this article we will discuss the role of cancer-associated inflammation and the particular role of MIF in malignant disease.
Collapse
Affiliation(s)
- Juliana Candido
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
61
|
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
|
62
|
Flores MBS, Rocha GZ, Damas-Souza DM, Osório-Costa F, Dias MM, Ropelle ER, Camargo JA, de Carvalho RB, Carvalho HF, Saad MJA, Carvalheira JBC. RETRACTED: Obesity-induced increase in tumor necrosis factor-α leads to development of colon cancer in mice. Gastroenterology 2012; 143:741-753.e4. [PMID: 22677195 DOI: 10.1053/j.gastro.2012.05.045] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 05/16/2012] [Accepted: 05/24/2012] [Indexed: 12/25/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief and Deputy Editor-in-Chief following an investigation into the data that were presented in several figures within the article. A number of images used in this article are believed to be duplicated images. The authors stated that they inadvertently inserted images of the wrong blots in several of the figures, resulting in the duplications; however, they did not address all of the concerns raised. Because the editors were no longer confident in the conclusions of the article based on these incorrect data, a decision was made to retract the paper. All authors have been notified of this decision. The University of Campinas (UNICAMP) in São Paulo, Brazil was contacted regarding these concerns, but to date the journal has received no response.
Collapse
Affiliation(s)
- Marcelo B S Flores
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Guilherme Z Rocha
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Danilo M Damas-Souza
- Department of Anatomy, Cell Biology, Physiology and Biophysics, State University of Campinas, Campinas, São Paulo, Brazil
| | - Felipe Osório-Costa
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Marília M Dias
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Eduardo R Ropelle
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Juliana A Camargo
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Rita B de Carvalho
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Hernandes F Carvalho
- Department of Anatomy, Cell Biology, Physiology and Biophysics, State University of Campinas, Campinas, São Paulo, Brazil
| | - Mario J A Saad
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - José B C Carvalheira
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
63
|
Ma X, Sun Y, Wang S, Yang Z, Song Y. Establishment of a malignant pleural effusion mouse model with lewis lung carcinoma cell lines expressing enhanced green fluorescent protein. Transl Lung Cancer Res 2012; 1:170-8. [PMID: 25806179 DOI: 10.3978/j.issn.2218-6751.2012.07.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 07/02/2012] [Indexed: 11/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Malignant pleural effusion (MPE) is a poor prognostic factor in patients with advanced lung cancer. The aim of this study is to establish a mouse model of MPE using Lewis lung carcinoma (LLC) cell lines expressing enhanced green fluorescent protein (EGFP). METHODS The mouse model was created by injecting LLC-EGFP cells directly into the pleural cavity of nude mice under the guidance of stereomicroscope and then mice were sacrificed periodically. The dynamic growth and metastasis of tumor cells were screened using in vivo fluorescence imaging. The remaining mice were subjected to transverse computed tomography (CT) periodically to analyze the rate of MPE formation. The survival rate and tumor metastasis were also observed after modeling. Pleural fluid was gently aspirated using a 1 mL syringe and its volume was measured. When two or more mice bore MPE at the same time, we calculated the average volume. The correlation of MPE with the integrated optical density (IOD) were analyzed. RESULTS Four days after the inoculation of LLC-EGFP cells, green fluorescence was observed by opening the chest wall. The tumor formation rate was 100%, and the IOD gradually increased after inoculation. The metastatic foci were mediastinal, contralateral pleural and pericardial. The metastasis rates were 87%, 73%, and 20%, respectively. CT imagings revealed that the rates of MPE formation on days 7, 14 and 21 were 13%, 46%, and 53%. The mean survival time of nude mice was 28.8 days. The average MPE volume increased obviously on day 10 and peaked on day 16 with a value of 0.5 mL. The MPE volume and IOD were significantly correlated (r=0.91, P<0.0001). CONCLUSIONS This study was the first to establish a mouse model of MPE by injecting LLC-EGFP into the pleural cavity under the guidance of a stereomicroscope. The model can enable dynamic observations of the biological behavior of tumor cells in the pleural cavity. It might be helpful for basic research on advanced lung cancer as well as anti-tumor drug development.
Collapse
Affiliation(s)
- Xingqun Ma
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Clinical School of the Medical College of Nanjing University, Nanjing 210002, China
| | - Yu Sun
- Nanjing Origin Center for Tumor Animal Model, Nanjing 210002, China
| | - Shouju Wang
- Department of Medical Imaging, Nanjing General Hospital of Nanjing Command, Clinical School of the Medical College of Nanjing University, Nanjing 210002, China
| | - Zhijian Yang
- Nanjing Origin Center for Tumor Animal Model, Nanjing 210002, China
| | - Yong Song
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Clinical School of the Medical College of Nanjing University, Nanjing 210002, China
| |
Collapse
|
64
|
Role of monocyte chemoattractant protein-1, tumor necrosis factor-alpha and interleukin-6 in the control of malignant pleural effusion and survival in patients with primary lung adenocarcinoma. Int J Biol Markers 2012; 27:e118-24. [PMID: 22505233 DOI: 10.5301/jbm.2012.9197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2012] [Indexed: 11/20/2022]
Abstract
This study aimed at assessing the role of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) in the control of pleural effusion (PE) and survival in patients with primary lung adenocarcinoma. The concentrations of the 3 cytokines were measured in PE from 79 lung adenocarcinoma patients with malignant pleural effusion (MPE) and 23 patients with tuberculosis. Data were correlated with the efficacy of MPE control and patient survival. The level of MCP-1 in PE was significantly higher in patients with lung adenocarcinoma than those with tuberculosis. By contrast, the levels of TNF-alpha and IL-6 were significantly lower in patients with lung adenocarcinoma than those with tuberculosis. An MCP-1 level greater than 3,187 pg/mL (which was used as a cutoff point) indicated failure to control MPE (odds ratio [OR]=2.82, 95% confidence interval [CI]=1.02-7.82, p=0.04). In multivariate analysis, MCP-1 was confirmed as an independent prognostic factor for progression-free survival (hazard ratio [HR]=2.02, 95% CI=1.24-3.30, p=0.01). The level of MCP-1 in PE appears to be a reliable surrogate marker for evaluating the therapeutic efficacy in the control of MPE and predicting survival in lung adenocarcinoma patients with MPE.
Collapse
|
65
|
Neutralization of tumor necrosis factor bioactivity ameliorates urethane-induced pulmonary oncogenesis in mice. Neoplasia 2012; 13:1143-51. [PMID: 22241960 DOI: 10.1593/neo.111224] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/12/2011] [Accepted: 10/17/2011] [Indexed: 01/08/2023] Open
Abstract
Tumor necrosis factor (TNF) has been implicated in inflammation-associated tumor progression. Although multiple reports identified a role for TNF signaling in established cancers, few studies have assessed the impact of TNF blockade on early tumor formation promotion. We aimed at exploring the effects of TNF neutralization in a preclinical mouse model of lung carcinogenesis. For this, Balb/c mice (n = 42) received four weekly intraperitoneal urethane injections (1 g/kg) and twice-weekly intraperitoneal soluble TNF receptor (etanercept; 10 mg/kg) administered during tumor initiation/promotion, tumor progression, or continuously (months 1, 6, and 1-8 after urethane start, respectively). Lung oncogenesis was assessed after 8 months. In separate short-term studies, Balb/c mice (n = 21) received a single control or urethane injection followed by twice-weekly intraperitoneal control or sTNFR:Fc injections. Lung inflammation was assessed after 1 week. We found that sTNFR:Fc treatment during tumor initiation/promotion resulted in a significant reduction of tumor number but not dimensions. However, sTNFR:Fc administered during tumor progression did not impact tumor multiplicity but significantly decreased tumor diameter. Continued sTNFR:Fc administration was effective in halting both respiratory tumor formation and progression in response to urethane. This favorable impact was associated with impaired cellular proliferation and new vessel formation in lung tumors. In addition, TNF neutralization altered the lung inflammatory response to urethane, evidenced by reductions in TNF and macrophage and increases in interferon γ and interleukin 10 content of the air spaces. sTNFR:Fc treatment of RAW264.7 macrophages downregulated TNF and enhanced interferon γ and interleukin 10 expression. In conclusion, TNF neutralization is effective against urethane-induced lung oncogenesis in mice and could present a lung chemoprevention strategy worth testing clinically.
Collapse
|
66
|
Psallidas I, Stathopoulos GT, Maniatis NA, Magkouta S, Moschos C, Karabela SP, Kollintza A, Simoes DCM, Kardara M, Vassiliou S, Papiris SA, Roussos C, Kalomenidis I. Secreted phosphoprotein-1 directly provokes vascular leakage to foster malignant pleural effusion. Oncogene 2012; 32:528-35. [PMID: 22370646 DOI: 10.1038/onc.2012.57] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Secreted phosphoprotein-1 (SPP1) promotes cancer cell survival and regulates tumor-associated angiogenesis and inflammation, both central to the pathogenesis of malignant pleural effusion (MPE). Here, we examined the impact of tumor- and host-derived SPP1 in MPE formation and explored the mechanisms by which the cytokine exerts its effects. We used a syngeneic murine model of lung adenocarcinoma-induced MPE. To dissect the effects of tumor- versus host-derived SPP1, we intrapleurally injected wild-type and SPP1-knockout C57/BL/6 mice with either wild-type or SPP1-deficient syngeneic lung cancer cells. We demonstrated that both tumor- and host-derived SPP1 promoted pleural fluid accumulation and tumor dissemination in a synergistic manner (P<0.001). SPP1 of host origin elicited macrophage recruitment into the cancer-affected pleural cavity and boosted tumor angiogenesis, whereas tumor-derived SPP1 curtailed cancer cell apoptosis in vivo. Moreover, the cytokine directly promoted vascular hyper-permeability independently of vascular endothelial growth factor. In addition, SPP1 of tumor and host origin differentially affected the expression of proinflammatory and angiogenic mediators in the tumor microenvironment. These results suggest that SPP1 of tumor and host origin impact distinct aspects of MPE pathobiology to synergistically promote pleural fluid formation and pleural tumor progression. SPP1 may present an attractive target of therapeutic interventions for patients with MPE.
Collapse
Affiliation(s)
- I Psallidas
- Marianthi Simou Laboratory, 1st Department of Critical Care & Pulmonary Services, Athens Medical School, Evangelismos Hospital, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Karabela SP, Psallidas I, Sherrill TP, Kairi CA, Zaynagetdinov R, Cheng DS, Vassiliou S, McMahon F, Gleaves LA, Han W, Stathopoulos I, Zakynthinos SG, Yull FE, Roussos C, Kalomenidis I, Blackwell TS, Stathopoulos GT. Opposing effects of bortezomib-induced nuclear factor-κB inhibition on chemical lung carcinogenesis. Carcinogenesis 2012; 33:859-67. [PMID: 22287559 DOI: 10.1093/carcin/bgs024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Since recent evidence indicates a requirement for epithelial nuclear factor (NF)-κB signaling in lung tumorigenesis, we investigated the impact of the NF-κB inhibitor bortezomib on lung tumor promotion and growth. We used an experimental model in which wild-type mice or mice expressing an NF-κB reporter received intraperitoneal urethane (1 g/kg) followed by twice weekly bortezomib (1 mg/kg) during distinct periods of tumor initiation/progression. Mice were serially assessed for lung NF-κB activation, inflammation and carcinogenesis. Short-term proteasome inhibition with bortezomib did not impact tumor formation but retarded the growth of established lung tumors in mice via effects on cell proliferation. In contrast, long-term treatment with bortezomib resulted in significantly increased lung tumor number and size. This tumor-promoting effect of prolonged bortezomib treatment was associated with perpetuation of urethane-induced inflammation and chronic upregulation of interleukin-1β and proinflammatory C-X-C motif chemokine ligands (CXCL) 1 and 2 in the lungs. In addition to airway epithelium, bortezomib inhibited NF-κB in pulmonary macrophages in vivo, presenting a possible mechanism of tumor amplification. In this regard, RAW264.7 macrophages exposed to bortezomib showed increased expression of interleukin-1β, CXCL1 and CXCL2. In conclusion, although short-term bortezomib may exert some beneficial effects, prolonged NF-κB inhibition accelerates chemical lung carcinogenesis by perpetuating carcinogen-induced inflammation. Inhibition of NF-κB in pulmonary macrophages appears to play an important role in this adverse process.
Collapse
Affiliation(s)
- Sophia P Karabela
- Applied Biomedical Research and Training Center Marianthi Simou, Department of Critical Care and Pulmonary Services, General Hospital Evangelismos, School of Medicine, National and Kapodistrian University of Athens, 10675 Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Hamsa TP, Kuttan G. Berberine inhibits pulmonary metastasis through down-regulation of MMP in metastatic B16F-10 melanoma cells. Phytother Res 2011; 26:568-78. [PMID: 21953764 DOI: 10.1002/ptr.3586] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 05/07/2011] [Accepted: 05/11/2011] [Indexed: 12/28/2022]
Abstract
The present study demonstrated the potential antimetastatic and antiinvasive effect of berberine using both in vivo mouse lung metastasis and in vitro models. Administration of berberine resulted in significant suppression of B16F-10 melanoma induced tumor nodule formation and enhanced the survival of tumor-bearing mice. Berberine treatment also decreased various biochemical parameters associated with lung metastasis. These inhibitory actions may be due to the significant suppression of several signaling molecules such as ERK1/2, NF-κB, ATF-2 and CREB involved in the transcription signaling pathways for MMP gene expression. It could also inhibit the migration and invasion of highly metastatic murine melanoma cells in a dose-dependent manner in vitro. The results clearly show that berberine could significantly inhibit experimental lung metastasis produced by intravenous injection of B16F-10 melanoma cells and this effect could be linked to the down-regulation of metastasis-related signaling molecules.
Collapse
Affiliation(s)
- T P Hamsa
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala, India
| | | |
Collapse
|
69
|
Abstract
Pleural malignancies, including primary malignant pleural mesothelioma and secondary pleural metastasis of various tumours resulting in malignant pleural effusion, are frequent and lethal diseases that deserve devoted translational research efforts for improvements to be introduced to the clinic. This paper highlights select clinical advances that have been accomplished recently and that are based on preclinical research on pleural malignancies. Examples are the establishment of folate antimetabolites in mesothelioma treatment, the use of PET in mesothelioma management and the discovery of mesothelin as a marker of mesothelioma. In addition to established translational advances, this text focuses on recent research findings that are anticipated to impact clinical pleural oncology in the near future. Such progress has been substantial, including the development of a genetic mouse model of mesothelioma and of transplantable models of pleural malignancies in immunocompetent hosts, the deployment of stereological and imaging methods for integral assessment of pleural tumour burden, as well as the discovery of the therapeutic potential of aminobiphosphonates, histone deacetylase inhibitors and ribonucleases against malignant pleural disease. Finally, key obstacles to overcome towards a more rapid advancement of translational research in pleural malignancies are outlined. These include the dissection of cell-autonomous and paracrine pathways of pleural tumour progression, the study of mesothelioma and malignant pleural effusion separately from other tumours at both the clinical and preclinical levels, and the expansion of tissue banks and consortia of clinical research of pleural malignancies.
Collapse
|
70
|
Daintain/AIF-1 promotes breast cancer cell migration by up-regulated TNF-α via activate p38 MAPK signaling pathway. Breast Cancer Res Treat 2011; 131:891-8. [PMID: 21509525 DOI: 10.1007/s10549-011-1519-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2011] [Accepted: 04/11/2011] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages can release a vast diversity of growth factors, proteolytic enzymes, cytokines, and inflammatory mediators. Many of these factors are key agents in cancer metastasis. Daintain/AIF-1 is a macrophage-derived inflammatory cytokine which defined a distinct subset of tumor-associated activated macrophages/microglial cells. Previous study demonstrated that daintain/AIF-1 could promote breast cancer proliferation through activating NF-κB/cyclin D1 pathway and facilitate tumor growth. However, the effect of Daintain/AIF-1 on cell migration and cancer metastasis has never been reported. Herein, we used a mimic tumor microenvironment by incubating breast cancer cell lines, MDA-MB-231 and MCF-7 cells, with macrophage-conditioned medium with or without purified daintain/AIF-1 polypeptide to evaluate cell migration. Results indicated that daintain/AIF-1 enhanced the migration of MDA-MB-231 and MCF-7 cells in the manner of TNF-α up-regulation. Further study found that daintain/AIF-1 activates p38 MAPK signaling pathway contributing to up-regulation of TNF-α in MDA-MB-231 and MCF-7 cells. Therefore, this novel daintain/AIF-1-p38-TNF-α pathway and insight into daintain/AIF-1 might have potential benefits in the control of tumor metastasis during cancer therapy.
Collapse
|
71
|
Takano T, Ohyama T, Kokumoto A, Satoh R, Hohdatsu T. Vascular endothelial growth factor (VEGF), produced by feline infectious peritonitis (FIP) virus-infected monocytes and macrophages, induces vascular permeability and effusion in cats with FIP. Virus Res 2011; 158:161-8. [PMID: 21473893 PMCID: PMC7114439 DOI: 10.1016/j.virusres.2011.03.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/18/2011] [Accepted: 03/25/2011] [Indexed: 12/07/2022]
Abstract
Feline infectious peritonitis virus (FIPV) causes a fatal disease called FIP in Felidae. The effusion in body cavity is commonly associated with FIP. However, the exact mechanism of accumulation of effusion remains unclear. We investigated vascular endothelial growth factor (VEGF) to examine the relationship between VEGF levels and the amounts of effusion in cats with FIP. Furthermore, we examined VEGF production in FIPV-infected monocytes/macrophages, and we used feline vascular endothelial cells to examine vascular permeability induced by the culture supernatant of FIPV-infected macrophages. In cats with FIP, the production of effusion was related with increasing plasma VEGF levels. In FIPV-infected monocytes/macrophages, the production of VEGF was associated with proliferation of virus. Furthermore, the culture supernatant of FIPV-infected macrophages induced hyperpermeability of feline vascular endothelial cells. It was suggested that vascular permeability factors, including VEGF, produced by FIPV-infected monocytes/macrophages might increase the vascular permeability and the amounts of effusion in cats with FIP.
Collapse
Affiliation(s)
- Tomomi Takano
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | | | | | | | | |
Collapse
|
72
|
Moschos C, Psallidas I, Cottin T, Kollintza A, Papiris S, Roussos C, Stathopoulos GT, Giannis A, Kalomenidis I. A sulindac analogue is effective against malignant pleural effusion in mice. Lung Cancer 2011; 73:171-5. [PMID: 21227533 DOI: 10.1016/j.lungcan.2010.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/27/2010] [Accepted: 12/06/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVES To examine whether a sulindac derivative (C-18) with previously reported anti-angiogenic properties limits malignant pleural effusion (MPE) formation in mice. METHODS MPE was generated by intrapleural injection of murine adenocarcinoma cells in C57BL/6 mice. Animals were divided into three groups, a control group and two treatment groups receiving intraperitoneally a daily dose of either 1 mg or 2 mg of C-18 for a total of 12 doses. Mice were sacrificed on day 14. MEASUREMENTS AND MAIN RESULTS Pleural fluid volume and the number of pleural tumor implantations were measured. Tumor angiogenesis, pleural vascular permeability and the host inflammatory response were also assessed. C-18 significantly limited pleural fluid formation and inhibited intrapleural tumor dissemination. The mean±SEM pleural fluid volume was 758±63 μl for the control group, compared to 492±120 μl (p=0.042) and 279±77 μl (p<0.001) for the low dose and high dose group of C-18, respectively. Control group animals had 6.2±1 intrapleural tumors, while C-18 treated animals had 3.1±0.8 (p=0.014) and 3±0.7 (p=0.009) for the low and high dose respectively. In addition C-18 significantly suppressed pleural vascular permeability. No significant difference in tumor angiogenesis and inflammatory response was observed, while there was also no measurable effect in tumor cell apoptosis and proliferation in vitro and in vivo. CONCLUSIONS C-18 halted experimental MPE formation and intrapleural tumor dissemination, through down-regulation of pleural vascular permeability.
Collapse
Affiliation(s)
- Charalampos Moschos
- Applied Biomedical Research & Training Center Marianthi Simou and George P. Livanos Laboratory, Athens, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Suspitsin EN, Levchenko EV, Moiseyenko FV, Ivantsov AO, Radzhabova SA, Matsko DE, Moiseyenko VM, Imyanitov EN. Rapid Symptomatic Improvement in Gefitinib-Treated Patients with EGFR-Mutated Lung Cancer: Possible Role of Downregulation of Inflammatory Molecules? ACTA ACUST UNITED AC 2011; 34:559-60. [DOI: 10.1159/000332123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
74
|
Stathopoulos GT, Sherrill TP, Karabela SP, Goleniewska K, Kalomenidis I, Roussos C, Fingleton B, Yull FE, Peebles RS, Blackwell TS. Host-derived interleukin-5 promotes adenocarcinoma-induced malignant pleural effusion. Am J Respir Crit Care Med 2010; 182:1273-81. [PMID: 20595227 PMCID: PMC3001265 DOI: 10.1164/rccm.201001-0001oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2010] [Accepted: 07/08/2010] [Indexed: 12/18/2022] Open
Abstract
RATIONALE IL-5 is a T helper 2 cytokine important in the trafficking and survival of eosinophils. Because eosinophils can be found in malignant pleural effusions (MPE) from mice and humans, we asked whether IL-5 is involved in the pathogenesis of MPE. OBJECTIVES To determine the role of IL-5 in MPE formation. METHODS The effects of IL-5 on experimental MPE induced in C57BL/6 mice by intrapleural injection of syngeneic lung (Lewis lung cancer [LLC]) or colon (MC38) adenocarcinoma cells were determined using wild-type (il5(+/+)) and IL-5-deficient (il5⁻(/)⁻) mice, exogenous administration of recombinant mouse (rm) IL-5, and in vivo antibody-mediated neutralization of endogenous IL-5. The direct effects of rmIL-5 on LLC cell proliferation and gene expression in vitro were determined by substrate reduction and microarray. MEASUREMENTS AND MAIN RESULTS Eosinophils and IL-5 were present in human and mouse MPE, but the cytokine was not detected in mouse (LLC) or human (A549) lung and mouse colon (MC38) adenocarcinoma-conditioned medium, suggesting production by host cells in MPE. Compared with il5(+/+) mice, il5⁻(/)⁻ mice showed markedly diminished MPE formation in response to both LLC and MC38 cells. Exogenous IL-5 promoted MPE formation in il5(+/+) and il5⁻(/)⁻ mice, whereas anti-IL-5 antibody treatment limited experimental MPE in il5(+/+) mice. Exogenous IL-5 had no effects on LLC cell proliferation and gene expression; however, IL-5 was found to be responsible for recruitment of eosinophils and tumor-promoting myeloid suppressor cells to MPE in vivo. CONCLUSIONS Host-derived IL-5 promotes experimental MPE and may be involved in the pathogenesis of human MPE.
Collapse
|
75
|
Mishra EK, Davies RJO. Advances in the investigation and treatment of pleural effusions. Expert Rev Respir Med 2010; 4:123-33. [PMID: 20387298 DOI: 10.1586/ers.09.67] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pleural effusions present a challenge for both diagnosis and treatment. They are a commonly presenting problem of a wide range of local and systemic potentially life threatening diseases and cause significant breathlessness. Significant advances have been made in the last 5 years in the diagnostic pathway and management options. This article reviews recent developments in the investigation of pleural effusions, particularly in pleural fluid analysis, biomarkers, imaging and pleural biopsy, and in the treatment of pleural infection and both malignant and benign effusions, including the use of indwelling pleural catheters. Although significant recent advances have been made in the management of pleural effusions, there the need still remains for further research if we are to reduce the morbidity and mortality caused by pleural effusions.
Collapse
Affiliation(s)
- Eleanor K Mishra
- Oxford Respiratory Trials Unit, Nuffield Department of Medicine, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK.
| | | |
Collapse
|
76
|
Psallidas I, Karabela SP, Moschos C, Sherrill TP, Kollintza A, Magkouta S, Theodoropoulou P, Roussos C, Blackwell TS, Kalomenidis I, Stathopoulos GT. Specific effects of bortezomib against experimental malignant pleural effusion: a preclinical study. Mol Cancer 2010; 9:56. [PMID: 20219102 PMCID: PMC2841124 DOI: 10.1186/1476-4598-9-56] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 03/10/2010] [Indexed: 12/23/2022] Open
Abstract
Background We have previously shown that nuclear factor (NF)-κB activation of mouse Lewis lung carcinoma (LLC) specifically promotes the induction of malignant pleural effusions (MPE) by these cells. In the present studies we hypothesized that treatment of immunocompetent mice with bortezomib tailored to inhibit cancer cell NF-κB activation and not proliferation specifically inhibits MPE formation by LLC cells. Results Treatment of LLC cells with low concentrations of bortezomib (100 ng/ml) inhibited NF-κB activation and NF-κB-dependent transcription, but not cellular proliferation. Bortezomib treatment of immunocompetent C57BL/6 mice bearing LLC-induced subcutaneous tumors and MPEs significantly blocked tumor-specific NF-κB activation. However, bortezomib treatment did not impair subcutaneous LLC tumor growth, but was effective in limiting LLC-induced MPE. This specific effect was evidenced by significant reductions in effusion accumulation and the associated mortality and was observed with both preventive (beginning before MPE formation) and therapeutic (beginning after MPE establishment) bortezomib treatment. The favorable impact of bortezomib on MPE was associated with suppression of cardinal MPE-associated phenomena, such as inflammation, vascular hyperpermeability, and angiogenesis. In this regard, therapeutic bortezomib treatment had identical favorable results on MPE compared with preventive treatment, indicating that the drug specifically counteracts effusion formation. Conclusions These studies indicate that proteasome inhibition tailored to block NF-κB activation of lung adenocarcinoma specifically targets the effusion-inducing phenotype of this tumor. Although the drug has limited activity against advanced solid lung cancer, it may prove beneficial for patients with MPE.
Collapse
Affiliation(s)
- Ioannis Psallidas
- Applied Biomedical Research & Training Center "Marianthi Simou", Department of Critical Care & Pulmonary Services, General Hospital "Evangelismos", National and Kapodistrian University of Athens, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Economidou F, Margaritopoulos G, Antoniou KM, Siafakas NM. The angiogenetic pathway in malignant pleural effusions: Pathogenetic and therapeutic implications. Exp Ther Med 2010; 1:3-7. [PMID: 23136584 DOI: 10.3892/etm_00000001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 10/06/2009] [Indexed: 01/09/2023] Open
Abstract
Increased permeability of the pleural microvasculature is generally attributed to the substances that are released in inflammatory and malignant pleural effusions, although the exact pathogenetic mechanisms of malignant pleural effusions are unclear. Current therapies used to prevent the re-accumulation of pleural fluid and relieve symptoms are of variable efficacy and may cause serious adverse effects. Understanding the mechanisms of fluid accumulation would hopefully permit the development of more specific, effective and safer treatment modalities. Angiogenesis, pleural vascular increased permeability and inflammation are considered central to the pathogenesis of malignant pleural effusions. Vascular endothelial growth factor (VEGF) is a member of the VEGF/platelet-derived factor gene family and consists of at least six isoforms. Since it was shown that VEGF contributes to the formation of malignant pleural effusions, there have been some attempts to implicate, therapeutically, this finding using different molecules (ZD6474, PTK 787 and bevacizumab). However, the role of the biological axis of VEGF and angiopoietins needs further investigation in both the pathogenesis and the treatment of malignant pleural effusion. In both non-small-cell lung carcinoma and breast cancer, it has been shown that the ligand for CXCR4, CXCL12 or SDF-1α, exhibited peak levels of expression in organs that were the preferred destination for their respective metastases. Recent findings imply that new therapeutic strategies aimed at blocking the SDF-1-CXCR4 axis may have significant applications for patients by modulating the trafficking of hemato/lymphopoietic cells and inhibiting the metastatic behavior of tumor cells as well. The purpose of this report is to review novel pathogenetic and therapeutic implications regarding the angiogenetic pathways in malignant pleural effusions.
Collapse
Affiliation(s)
- Foteini Economidou
- Department of Thoracic Medicine, Medical School, University of Crete, Heraklion, Greece
| | | | | | | |
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW Malignant pleural effusion (MPE) poses a common and significant clinical problem. Its pathogenesis is poorly understood and therapeutic options are limited. Herein are summarized animal models of MPE and their contributions in unveiling new aspects of the pathobiology of the condition. RECENT FINDINGS In recent years, different groups have developed novel models of MPE, including a genetic mouse model of spontaneous mesothelioma development, a model of adenocarcinoma-induced MPE in immunocompetent mice, as well as models of human cancer-induced MPE in immunocompromised animals, all relevant to the human condition to a different extent. Work using these models has yielded novel insights into the pathogenesis of mesothelioma as well as into the mechanisms of intrapleural malignant effusion accumulation and tumor dissemination. The data produced underline the significance of tumor-associated inflammation, angiogenesis, and vascular hyperpermeability in the pathogenesis of MPE. SUMMARY In the past few years, novel approaches to induce experimental MPE have yielded new insights into its pathogenesis and have identified possible therapeutic targets to block pleural fluid exudation induced by primary and metastatic cancer cells in the pleural space.
Collapse
|
79
|
Charles KA, Kulbe H, Soper R, Escorcio-Correia M, Lawrence T, Schultheis A, Chakravarty P, Thompson RG, Kollias G, Smyth JF, Balkwill FR, Hagemann T. The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest 2009; 119:3011-23. [PMID: 19741298 DOI: 10.1172/jci39065] [Citation(s) in RCA: 253] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2009] [Accepted: 07/15/2009] [Indexed: 12/29/2022] Open
Abstract
Cytokines orchestrate the tumor-promoting interplay between malignant cells and the immune system. In many experimental and human cancers, the cytokine TNF-alpha is an important component of this interplay, but its effects are pleiotropic and therefore remain to be completely defined. Using a mouse model of ovarian cancer in which either TNF receptor 1 (TNFR1) signaling was manipulated in different leukocyte populations or TNF-alpha was neutralized by antibody treatment, we found that this inflammatory cytokine maintained TNFR1-dependent IL-17 production by CD4+ cells and that this led to myeloid cell recruitment into the tumor microenvironment and enhanced tumor growth. Consistent with this, in patients with advanced cancer, treatment with the TNF-alpha-specific antibody infliximab substantially reduced plasma IL-17 levels. Furthermore, expression of IL-1R and IL-23R was downregulated in CD4+CD25- cells isolated from ascites of ovarian cancer patients treated with infliximab. We have also shown that genes ascribed to the Th17 pathway map closely with the TNF-alpha signaling pathway in ovarian cancer biopsy samples, showing particularly high levels of expression of genes encoding IL-23, components of the NF-kappaB system, TGF-beta1, and proteins involved in neutrophil activation. We conclude that chronic production of TNF-alpha in the tumor microenvironment increases myeloid cell recruitment in an IL-17-dependent manner that contributes to the tumor-promoting action of this proinflammatory cytokine.
Collapse
Affiliation(s)
- Kellie A Charles
- Centre for Cancer and Inflammation, Institute of Cancer, Barts and The London School of Medicine and Dentistry,Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Okayama T, Kokura S, Ishikawa T, Adachi S, Hattori T, Takagi T, Handa O, Naito Y, Yoshikawa T. Antitumor effect of pretreatment for colon cancer cells with hyperthermia plus geranylgeranylacetone in experimental metastasis models and a subcutaneous tumor model of colon cancer in mice. Int J Hyperthermia 2009; 25:141-9. [PMID: 19337914 DOI: 10.1080/02656730802631783] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE We examined whether hyperthermia attenuated the metastatic potential of colon cancer through the induction of heat shock protein 70 (Hsp70). MATERIALS AND METHODS Colon26 cells were separated into four groups: (1) no pretreatment, (2) hyperthermia at 42 degrees C for 1 hour, (3) pretreatment with geranylgeranylacetone (GGA) 10(-6) M for 2 hours, and (4) hyperthermia after GGA treatment. We measured cell viabilities and the contents of Hsp70. We assessed nuclear factor-kappa-B (NF-kappa-B) status with and without tumor necrosis factor-alpha (TNF-alpha) stimulation. For in vivo study, colon26 cells were injected via the tail vein or into a subcutaneous area of mice and the numbers of lung metastatic nodules or the volumes of subcutaneous tumors were assessed. Untreated cells were incubated with PKH26. Experimental metastasis models were then generated and used to assess the fixed cancer cells. RESULTS Tumor development in the subcutaneous tumor models and cell viabilities were similar among the four groups. However, the GGA plus hyperthermia group had fewer lung metastatic nodules in the experimental lung metastasis model and higher Hsp70 induction than the other cell groups. The GGA plus hyperthermia pretreatment group also showed a lower number of fixed cells in lungs and lower activation of NF-kappa-B by TNF-alpha than the other cell groups. CONCLUSIONS It is suggested the metastatic potential but not the proliferation potency of cancer cells is inhibited by the transient induction of Hsp70.
Collapse
Affiliation(s)
- Tetsuya Okayama
- Department of Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
Tumour necrosis factor (TNF) is a major inflammatory cytokine that was first identified for its ability to induce rapid haemorrhagic necrosis of experimental cancers. When efforts to harness this anti-tumour activity in cancer treatments were underway, a paradoxical tumour-promoting role of TNF became apparent. Now that links between inflammation and cancer are appreciated, is TNF a target or a therapeutic in malignant disease -- or both?
Collapse
Affiliation(s)
- Frances Balkwill
- Centre for Cancer and Inflammation, Institute of Cancer, Barts, UK.
| |
Collapse
|
82
|
Zhang J, Peng B. NF-kappaB promotes iNOS and VEGF expression in salivary gland adenoid cystic carcinoma cells and enhances endothelial cell motility in vitro. Cell Prolif 2009; 42:150-61. [PMID: 19317804 DOI: 10.1111/j.1365-2184.2009.00588.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Tumour cell-derived angiogenic-related factors and endothelial cell mobility are essential for neovascularization and haematogenous metastasis of adenoid cystic carcinoma (ACC) of the human salivary glands. Our previous study demonstrated that vascular endothelial growth factor (VEGF), inducible nitric oxide synthase (iNOS), and nuclear factor kappaB (NF-kappaB) staining correlated with ACC microvessel density. However, there still remains a lack of direct evidence to clarify the function of liposaccharide-induced NF-kappaB activity in ACC angiogenesis. OBJECTIVES This study aimed to demonstrate the function of liposaccharide-induced NF-kappaB signalling pathway (which would be relevant to angiogenesis) of ACC cell lines. MATERIALS AND METHODS A PCMV-IkappaBalphaM vector transfection assay was performed to inhibit NF-kappaB activity. Constitutive and liposaccharide-induced NF-kappaB activity, along with VEGF and iNOS expression, was detected by electrophoretic mobility shift assays, reverse transcription-polymerase chain reaction and Western blot assay. Tumour cell-derived NO(2) (-) was evaluated by a nitrite determination assay, and endothelial cell mobility was investigated by endothelial cell proliferation, migration and tube formation assays. RESULTS We demonstrated that regulation of VEGF and iNOS expression utilized the NF-kappaB signalling pathway in ACC cell lines. In the constitutive and liposaccharide-induced condition, NF-kappaB activity was blocked by phosphorylation-defective IkappaBalpha. Accordingly, mRNA and protein expression of VEGF and iNOS were significantly decreased, with a great drop in endogenous NO(2) (-). Furthermore, supernatant from ACC-2/IkBaM and ACC-M/IkBaM cells resulted in much lower endothelial cell mobility than that from ACC-2 and ACC-M cells. CONCLUSIONS Our results suggest that NF-kappaB promotes iNOS and VEGF expression in salivary gland ACC cells and enhances endothelial cell motility in vitro.
Collapse
Affiliation(s)
- Jiali Zhang
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, Wuhan University, Wuhan, China
| | | |
Collapse
|
83
|
Moschos C, Psallidas I, Kollintza A, Karabela S, Papapetropoulos A, Papiris S, Light RW, Roussos C, Stathopoulos GT, Kalomenidis I. The angiopoietin/Tie2 axis mediates malignant pleural effusion formation. Neoplasia 2009; 11:298-304. [PMID: 19242611 PMCID: PMC2647732 DOI: 10.1593/neo.81480] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 01/14/2009] [Accepted: 01/14/2009] [Indexed: 11/18/2022]
Abstract
PURPOSE Angiopoietins and their receptor, Tie2, participate in angiogenesis, regulation of vascular permeability, and inflammation, all central to the pathogenesis of malignant pleural effusions (MPEs). In the present study, we aimed to examine the role of the angiopoietin/Tie2 axis in MPE pathogenesis. EXPERIMENTAL DESIGN MPE was induced by intrapleural injection of murine adenocarcinoma cells in C57BL/6 mice. Animals were given twice-weekly intraperitoneal injections of 40 mg/kg MuTekdeltaFc or vehicle. MuTekdeltaFc is a soluble Tie2 (sTie2) receptor that binds murine angiopoietins thereby disrupting their interaction with Tie2 receptors expressed on tissues. Animals were killed on day 14. RESULTS Angiopoietin/Tie2 axis blockade significantly reduced pleural fluid volume and pleural tumor foci. The mean +/- SEM pleural fluid volumes were 617 +/- 48 microl and 316 +/- 62 microl for the control and treated groups, respectively (P = .001), whereas the mean +/- SEM tumor foci were 7.3 +/- 1.0 and 3.0 +/- 0.52 for the control and treated groups, respectively (P = .001). In addition, tumor-associated cachexia, tumor angiogenesis, pleural vascular permeability, recruitment of inflammatory cells to the pleural cavity, and local elaboration of vascular endothelial growth factor and interleukin 6 were also downregulated, and tumor cell apoptosis was induced in animals treated with the inhibitor. CONCLUSIONS Our results indicate that the angiopoietin/Tie2 axis is an important component of MPE pathogenesis. Further studies are required to determine whether therapeutic interventions targeting this pathway could be beneficial for patients with MPE.
Collapse
Affiliation(s)
- Charalampos Moschos
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- Department of Critical Care & Pulmonary Services, General Hospital “Evangelismos,” School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Psallidas
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- 2nd Department of Pulmonary Medicine, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Androniki Kollintza
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
| | - Sophia Karabela
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
| | | | - Spyros Papiris
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- 2nd Department of Pulmonary Medicine, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Richard W Light
- Division of Allergy, Pulmonary, & Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Charis Roussos
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- Department of Critical Care & Pulmonary Services, General Hospital “Evangelismos,” School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios T Stathopoulos
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- Department of Critical Care & Pulmonary Services, General Hospital “Evangelismos,” School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Kalomenidis
- Applied Biomedical Research & Training Center “Marianthi Simou” and “George P. Livanos” Laboratory, Athens, Greece
- 2nd Department of Pulmonary Medicine, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
84
|
Therapeutic effects of recombinant human endostatin adenovirus in a mouse model of malignant pleural effusion. J Cancer Res Clin Oncol 2009; 135:1149-57. [PMID: 19219619 DOI: 10.1007/s00432-009-0555-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 01/22/2009] [Indexed: 02/05/2023]
Abstract
PURPOSE Malignant pleural effusion (MPE) is a common clinical problem in patients with advanced cancer. Evidence suggests that tumor-mediated angiogenesis and enhanced vascular permeability in the pleural wall are due to high levels of vascular endothelial growth factor (VEGF), which plays an important role in the pathogenesis of MPE. The present study was designed to test whether the recombinant adenovirus-mediated delivery of human endostatin (Ad-hEndo), one of the potent inhibitors of angiogenesis, would inhibit the formation and progression of MPE. METHODS We developed a novel mouse model of MPE by injecting Lewis lung carcinoma (LLC) cells directly into pleural cavity of C57BL/6 mice. To evaluate the therapeutic effects of endostatin in this MPE model, we injected the Ad-hEndo into the pleural cavity of MPE-bearing mice three times with the 3-day interval. RESULTS We found that this treatment resulted in significant reduction in pleural effusion volume, the number of pleural tumor foci, microvessel density, and vascular permeability, while it significantly prolonged the survival time. In addition, VEGF level of MPE in the group administered with the Ad-hEndo was obviously decreased as compared with that in the two control groups administered with null-adenovirus (Ad-null) or normal saline. CONCLUSIONS Our work provides a rationale for future studies toward evaluating the effectiveness of the adenovirus-based endostatin therapy for MPE.
Collapse
|
85
|
Abstract
Tumor necrosis factor (TNF) is a multifunctional cytokine that plays important roles in diverse cellular events such as cell survival, proliferation, differentiation, and death. As a pro-inflammatory cytokine, TNF is secreted by inflammatory cells, which may be involved in inflammation-associated carcinogenesis. TNF exerts its biological functions through activating distinct signaling pathways such as nuclear factor-kappaB (NF-kappaB) and c-Jun N-terminal kinase (JNK). NF-kappaB is a major cell survival signal that is anti-apoptotic, whereas sustained JNK activation contributes to cell death. The crosstalk between the NF-kappaB and JNK is involved in determining cellular outcomes in response to TNF. In regard to cancer, TNF is a double-dealer. On one hand, TNF could be an endogenous tumor promoter, because TNF stimulates the growth, proliferation, invasion and metastasis, and tumor angiogenesis of cancer cells. On the other hand, TNF could be a cancer killer. The property of TNF in inducing cancer cell death renders it a potential cancer therapeutic, although much work is needed to reduce its toxicity for systematic TNF administration. Recent studies have focused on sensitizing cancer cells to TNF-induced apoptosis through inhibiting survival signals such as NF-kappaB, by combined therapy. In this article we provide an overview of the roles of TNF-induced signaling pathways in cancer biology with specific emphasis on carcinogenesis and cancer therapy.
Collapse
Affiliation(s)
- Xia Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | | |
Collapse
|
86
|
Stathopoulos GT, Psallidas I, Moustaki A, Moschos C, Kollintza A, Karabela S, Porfyridis I, Vassiliou S, Karatza M, Zhou Z, Joo M, Blackwell TS, Roussos C, Graf D, Kalomenidis I. A central role for tumor-derived monocyte chemoattractant protein-1 in malignant pleural effusion. J Natl Cancer Inst 2008; 100:1464-76. [PMID: 18840818 DOI: 10.1093/jnci/djn325] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Tumor cells in malignant pleural effusions (MPEs) are an important source of monocyte chemoattractant protein (MCP)-1. However, the role of tumor-derived MCP-1 in the pathogenesis and progression of MPE has not been determined. METHODS B16 mouse skin melanoma cells, which are deficient in MCP-1 expression, and mouse Lewis lung cancer (LLC) cells, which express high levels of MCP-1, were engineered to stably express MCP-1 and short hairpin RNAs (shRNAs) targeting the MCP-1 transcript, respectively. Cells were injected into the pleural cavities of syngeneic immunocompetent mice, and MPE volume and pleural tumors were quantified at necropsy (day 14). MCP-1 and other mediators were determined by cytometric bead array and enzyme-linked immunosorbent assay, and mononuclear and endothelial cells were identified by immunolabeling of F4/80 and factor VIII-related antigen respectively. Mouse survival was assessed using Kaplan-Meier analysis. Vascular permeability in mice with MPE was assessed using albumin-binding Evans blue. Statistical tests were two-sided. RESULTS LLC cells expressing shRNA against MCP-1 elaborated less than 5% of the MCP-1 level in cells expressing nonspecific shRNA (control cells), and intrapleural delivery of these cells resulted in less MPE (mean MPE volume = 86 and 585 muL, respectively; difference = 499 muL; 95% confidence interval [CI] = 331 to 669 muL; P < .001), reduced MCP-1 levels in the pleural fluid, and lower mortality than when control cells were delivered. Overexpression of MCP-1 in intrapleurally injected B16 melanoma cells led to increased MPE and reduced survival. In mice with MPE, MCP-1 was a potent inducer of vascular permeability, mononuclear recruitment, and, in pleural tumors, of angiogenesis. CONCLUSION MCP-1 produced by tumor cells is an important determinant of their capacity to induce the formation of MPE and may be a useful target for the treatment of malignant pleural disease.
Collapse
Affiliation(s)
- Georgios T Stathopoulos
- Applied Biomedical Research and Training Center Marianthi Simou, Department of Critical Care and Pulmonary Services, General Hospital Evangelismos, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Stathopoulos GT, Moschos C, Loutrari H, Kollintza A, Psallidas I, Karabela S, Magkouta S, Zhou Z, Papiris SA, Roussos C, Kalomenidis I. Zoledronic acid is effective against experimental malignant pleural effusion. Am J Respir Crit Care Med 2008; 178:50-9. [PMID: 18388351 DOI: 10.1164/rccm.200710-1513oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Aminobiphosphonates, such as zoledronic acid (ZA), exert potent indirect antitumor effects and are currently being tested against human solid tumors. The antitumor actions of aminobiphosphonates, including angiostasis, are relevant to the pathogenesis of malignant pleural effusion (MPE), but no study has addressed the efficacy of these compounds against malignant pleural disease. OBJECTIVES Here we hypothesized that treatment of immunocompetent mice with ZA would halt tumor progression in a mouse model of adenocarcinoma-induced MPE. METHODS To induce MPE in mice, Lewis lung carcinoma cells were delivered directly into the pleural space. Subsequently, animals were treated with ZA in both a prevention and a regression protocol. MEASUREMENTS AND MAIN RESULTS ZA treatment resulted in significant reductions in pleural fluid accumulation and tumor dissemination, while it significantly prolonged survival. These effects of ZA were linked to enhanced apoptosis of pleural tumor cells, decreased formation of new vessels in pleural tumors, and reduced pleural vascular permeability. In addition, ZA was able to inhibit the recruitment of mononuclear cells to pleural tumors, with concomitant reductions in matrix metalloproteinase-9 release into the pleural space. Finally, ZA limited the expression of proinflammatory and angiogenic mediators, as well as the activity of small GTP proteins Ras and RhoA, in tumor cells in vivo and in vitro. CONCLUSIONS ZA is effective against experimental MPE, suggesting that this intervention should be considered for testing in clinical trials.
Collapse
Affiliation(s)
- Georgios T Stathopoulos
- Department of Critical Care and Pulmonary Services, Marianthi Simou and George P. Livanos Laboratory, General Hospital Evangelismos, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
|