1
|
Al-Azzawi HMA, Hamza SA, Paolini R, Arshad F, Patini R, O'Reilly L, McCullough M, Celentano A. Towards an emerging role for anticoagulants in cancer therapy: a systematic review and meta-analysis. FRONTIERS IN ORAL HEALTH 2024; 5:1495942. [PMID: 39568788 PMCID: PMC11576436 DOI: 10.3389/froh.2024.1495942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024] Open
Abstract
Background Anticoagulants, renowned for their role in preventing blood clot formation, have captivated researchers' attention for the exploitation of their potential to inhibit cancer in pre-clinical models. Objectives To undertake a systematic review and meta-analysis of the effects of anticoagulants in murine cancer research models. Further, to present a reference tool for anticoagulant therapeutic modalities relating to future animal pre-clinical models of cancer and their translation into the clinic. Methods Four databases were utilized including Medline (Ovid), Embase (Ovid), Web of science, and Scopus databases. We included studies relating to any cancer conducted in murine models that assessed the effect of traditional anticoagulants (heparin and its derivatives and warfarin) and newer oral anticoagulants on cancer. Results A total of 6,158 articles were identified in an initial multi-database search. A total of 157 records were finally included for data extraction. Studies on heparin species and warfarin demonstrated statistically significant results in favour of tumour growth and metastasis inhibition. Conclusion Our findings constitute a valuable reference guide for the application of anticoagulants in cancer research and explore the promising utilization of non-anticoagulants heparin in preclinical cancer research. Systematic Review Registration PROSPERO [CRD42024555603].
Collapse
Affiliation(s)
| | - Syed Ameer Hamza
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Fizza Arshad
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Romeo Patini
- Head and Neck Department, "Fondazione Policlinico Universitario A. Gemelli-IRCCS" School of Dentistry, Catholic University of Sacred Heart-Rome Largo A. Gemelli, Rome, Italy
| | - Lorraine O'Reilly
- Clinical Translation Centre, Cancer Biology and Stem Cells Division and Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
2
|
Al-Azzawi HMA, Hamza SA, Lin Z, Paolini R, McCullough M, Yap T, Celentano A. Potential effects of anticoagulants in preclinical mice models of oral cancer: A systematic review. Oral Dis 2024; 30:962-965. [PMID: 36825396 DOI: 10.1111/odi.14546] [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: 02/03/2023] [Revised: 02/12/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common head and neck cancer. There is mounting evidence to suggest that several components of the coagulation system directly affect carcinogenesis. Our recent in vitro studies demonstrated, for the first time, that various anticoagulants have anticancer effects on OSCC. They also showed the need for the immediate translation of these experimental conditions from bench to preclinical animal models. Here, we carried out a systematic review to summarise existing evidence on murine models built around the interactions between anticoagulants and oral cancer. Only one preclinical murine study was included in our systematic review, investigating the role of heparins in tumour pathophysiology. The paucity of evidence regarding the interactions between oral squamous cell carcinoma and anticoagulants emphasises the urgency with which further preclinical research should be conducted.
Collapse
Affiliation(s)
| | - Syed Ameer Hamza
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Zichen Lin
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Tami Yap
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
3
|
Paillasse MR, Esquerré M, Bertrand FA, Poussereau-Pomié C, Pichery M, Visentin V, Gueguen-Dorbes G, Gaujarengues F, Barron P, Badet G, Briaux A, Ancey PB, Sibrac D, Erdociain E, Özcelik D, Meneyrol J, Martin V, Gomez-Brouchet A, Selves J, Rochaix P, Battistella M, Lebbé C, Delord JP, Dol-Gleizes F, Bono F, Blanc I, Alam A, Hunneyball I, Whittaker M, Fons P. Targeting Tumor Angiogenesis with the Selective VEGFR-3 Inhibitor EVT801 in Combination with Cancer Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2022; 2:1504-1519. [PMID: 36970050 PMCID: PMC10035370 DOI: 10.1158/2767-9764.crc-22-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/13/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022]
Abstract
The receptor tyrosine kinase VEGFR-3 plays a crucial role in cancer-induced angiogenesis and lymphangiogenesis, promoting tumor development and metastasis. Here, we report the novel VEGFR-3 inhibitor EVT801 that presents a more selective and less toxic profile than two major inhibitors of VEGFRs (i.e., sorafenib and pazopanib). As monotherapy, EVT801 showed a potent antitumor effect in VEGFR-3–positive tumors, and in tumors with VEGFR-3–positive microenvironments. EVT801 suppressed VEGF-C–induced human endothelial cell proliferation in vitro and tumor (lymph)angiogenesis in different tumor mouse models. In addition to reduced tumor growth, EVT801 decreased tumor hypoxia, favored sustained tumor blood vessel homogenization (i.e., leaving fewer and overall larger vessels), and reduced important immunosuppressive cytokines (CCL4, CCL5) and myeloid-derived suppressor cells (MDSC) in circulation. Furthermore, in carcinoma mouse models, the combination of EVT801 with immune checkpoint therapy (ICT) yielded superior outcomes to either single treatment. Moreover, tumor growth inhibition was inversely correlated with levels of CCL4, CCL5, and MDSCs after treatment with EVT801, either alone or combined with ICT. Taken together, EVT801 represents a promising anti(lymph)angiogenic drug for improving ICT response rates in patients with VEGFR-3 positive tumors.
Significance:
The VEGFR-3 inhibitor EVT801 demonstrates superior selectivity and toxicity profile than other VEGFR-3 tyrosine kinase inhibitors. EVT801 showed potent antitumor effects in VEGFR-3–positive tumors, and tumors with VEGFR-3–positive microenvironments through blood vessel homogenization, and reduction of tumor hypoxia and limited immunosuppression. EVT801 increases immune checkpoint inhibitors’ antitumor effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Gaelle Badet
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | - Anne Briaux
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | | | - David Sibrac
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | | | | | | | | | - Anne Gomez-Brouchet
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Janik Selves
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Philippe Rochaix
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Maxime Battistella
- 5Université de Paris, Department of Pathology, AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Céleste Lebbé
- 6Université de Paris, Department of Dermatology, AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Jean-Pierre Delord
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | | | | | | | | | | | | | - Pierre Fons
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| |
Collapse
|
4
|
Aqueous outflow channels and its lymphatic association: A review. Surv Ophthalmol 2021; 67:659-674. [PMID: 34656556 PMCID: PMC9008077 DOI: 10.1016/j.survophthal.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 11/24/2022]
Abstract
The human eye has a unique immune architecture and behavior. While the conjunctiva is known to have a well-defined lymphatic drainage system, the cornea, sclera, and uveal tissues were historically considered "alymphatic" and thought to be immune privileged. The very fact that the aqueous outflow channels carry a clear fluid (aqueous humor) along the outflow pathway makes it hard to ignore its lymphatic-like characteristics. The development of novel lymphatic lineage markers and expression of these markers in aqueous outflow channels and improved imaging capabilities has sparked a renewed interest in the study of ocular lymphatics. Ophthalmic lymphatic research has had a directional shift over the last decade, offering an exciting new physiological platform that needs further in-depth understanding. The evidence of a presence of distinct lymphatic channels in the human ciliary body is gaining significant traction. The uveolymphatic pathway is an alternative new route for aqueous outflow and adds a new dimension to pathophysiology and management of glaucoma. Developing novel animal models, markers, and non-invasive imaging tools to delineate the core anatomical structure and physiological functions may help pave some crucial pathways to understand disease pathophysiology and help develop novel targeted therapeutic approaches for glaucoma.
Collapse
|
5
|
Ma SN, Mao ZX, Wu Y, Liang MX, Wang DD, Chen X, Chang PA, Zhang W, Tang JH. The anti-cancer properties of heparin and its derivatives: a review and prospect. Cell Adh Migr 2021; 14:118-128. [PMID: 32538273 PMCID: PMC7513850 DOI: 10.1080/19336918.2020.1767489] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heparin, including unfractionated heparin (UFH), low-molecular-weight heparin (LMWH) and heparin derivatives, are commonly used in venous thromboembolism treatment and reportedly have beneficial effects on cancer survival. Heparin can affect the proliferation, adhesion, angiogenesis, migration and invasion of cancer cells via multiple mechanisms. The main mechanisms involve inhibition of heparanase, P-/L-selectin, angiogenesis, and interference with the CXCL12-CXCR4 axis. Here we summarize the current experimental evidence regarding the anti-cancer role of heparin and its derivatives, and conclude that there is evidence to support heparin’s role in inhibiting cancer progression, making it a promising anti-cancer agent.
Collapse
Affiliation(s)
- Sai-Nan Ma
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China.,Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University , Suqian, P.R.China
| | - Zhi-Xiang Mao
- Department of Oncology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, P.R. China
| | - Yang Wu
- Core Facility, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Ming-Xing Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Dan-Dan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Xiu Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Ping-An Chang
- Urinary Surgery, Dongtai People's Hospital , Dongtai, P.R. China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| |
Collapse
|
6
|
Manspeaker MP, Thomas SN. Lymphatic immunomodulation using engineered drug delivery systems for cancer immunotherapy. Adv Drug Deliv Rev 2020; 160:19-35. [PMID: 33058931 PMCID: PMC7736326 DOI: 10.1016/j.addr.2020.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Though immunotherapy has revolutionized the treatment of cancer to improve disease outcomes, an array of challenges remain that limit wider clinical success, including low rate of response and immune-related adverse events. Targeting immunomodulatory drugs to therapeutically relevant tissues offers a way to overcome these challenges by potentially enabling enhanced therapeutic efficacy and decreased incidence of side effects. Research highlighting the importance of lymphatic tissues in the response to immunotherapy has increased interest in the application of engineered drug delivery systems (DDSs) to enable specific targeting of immunomodulators to lymphatic tissues and cells that they house. To this end, a variety of DDS platforms have been developed that enable more efficient uptake into lymphatic vessels and lymph nodes to provide targeted modulation of the immune response to cancer. This can occur either by delivery of immunotherapeutics to lymphatics tissues or by direct modulation of the lymphatic vasculature itself due to their direct involvement in tumor immune processes. This review will highlight DDS platforms that, by enabling the activities of cancer vaccines, chemotherapeutics, immune checkpoint blockade (ICB) antibodies, and anti- or pro-lymphangiogenic factors to lymphatic tissues through directed delivery and controlled release, augment cancer immunotherapy.
Collapse
Affiliation(s)
- Margaret P Manspeaker
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
7
|
Shibata MA, Shibata E, Tanaka Y, Shiraoka C, Kondo Y. Soluble Vegfr3 gene therapy suppresses multi-organ metastasis in a mouse mammary cancer model. Cancer Sci 2020; 111:2837-2849. [PMID: 32539229 PMCID: PMC7419054 DOI: 10.1111/cas.14531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence on the association of VEGF-C with lymphangiogenesis and lymph node metastasis implicates lymphatic vessels as a potential target in anti-cancer therapy. To evaluate whether blocking VEGF-C and VEGFR-3 signaling can inhibit multi-organ metastases, a mouse metastatic mammary cancer model was subjected to gene therapy using a soluble VEGFR-3 expression vector (psVEGFR-3). We showed that psVEGFR-3 significantly diminished cell growth in vitro with or without added VEGF-C, and significantly reduced primary tumor growth and tumor metastases to wide-spectrum organs in vivo. Although apoptotic cell death and angiogenesis levels did not differ between the control and psVEGFR-3 groups, cell proliferation and lymphangiogenesis in the mammary tumors were significantly decreased in the psVEGFR-3 group. Furthermore, lymphatic vessel invasion was significantly inhibited in this group. Real-time RT-PCR analysis revealed significantly high expression of the Vegfr3 gene due to gene therapy, and the transcriptional levels of Pcna and Lyve1 tended to decrease in the psVEGFR-3 group. Immunofluorescence staining indicated that phospho-tyrosine expression was considerably lower in tumor cells of psVEGFR-3-treated mammary carcinomas than those of control tumors. Double immunofluorescence staining indicated that phospho-tyrosine+ /LYVE-1+ (a lymphatic vessel marker) tended to decrease in psVEGFR-3-treated mammary carcinomas compared with control mice, indicating a decline in the activity of the VEGF-C/VEGFR-3 axis. These findings showed that a blockade of VEGF-C/VEGFR-3 signaling caused by sVEGFR-3 sequestered VEGF-C and prevented the side-effects of anti-angiogenesis and suppressed overall metastases, suggesting their high clinical significance.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Eiko Shibata
- Department of Molecular Innovation in Lipidology, National Cerebral & Cardiovascular Center Research Institute, Suita, Japan
| | - Yoshihisa Tanaka
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Chinatsu Shiraoka
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Yoichi Kondo
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
8
|
Wang H, Chen Y, Li W, Sun L, Chen H, Yang Q, Zhang H, Zhang W, Yuan H, Zhang H, Xing L, Sun W. Effect of VEGFC on lymph flow and inflammation-induced alveolar bone loss. J Pathol 2020; 251:323-335. [PMID: 32418202 PMCID: PMC10587832 DOI: 10.1002/path.5456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/21/2022]
Abstract
The lymphatic system plays a crucial role in the maintenance of tissue fluid homeostasis and the immunological response to inflammation. The effects of lymphatic drainage dysfunction on periodontitis have not been well studied. Here we show that lymphatic vessel endothelial receptor 1 (LYVE1)+ /podoplanin (PDPN)+ lymphatic vessels (LVs) are increased in the periodontal tissues, with accumulation close to the alveolar bone surface, in two murine periodontitis models: rheumatoid arthritis (RA)-associated periodontitis and ligature-induced periodontitis. Further, PDPN+ /alpha-smooth muscle actin (αSMA)- lymphatic capillaries are increased, whereas PDPN+ /αSMA+ collecting LVs are decreased significantly in the inflamed periodontal tissues. Both mouse models of periodontitis have delayed lymph flow in periodontal tissues, increased TRAP-positive osteoclasts, and significant alveolar bone loss. Importantly, the local administration of adeno-associated virus for vascular endothelial growth factor C, the major growth factor that promotes lymphangiogenesis, increases the area and number of PDPN+ /αSMA+ collecting LVs, promotes local lymphatic drainage, and reduces alveolar bone loss in both models of periodontitis. Lastly, LYVE1+ /αSMA- lymphatic capillaries are increased, whereas LYVE1+ /αSMA+ collecting LVs are decreased significantly in gingival tissues of patients with chronic periodontitis compared with those of clinically healthy controls. Thus, our findings reveal an important role of local lymphatic drainage in periodontal inflammation-mediated alveolar bone loss. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Yuyi Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Wenlei Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Lian Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Hongyu Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Qiudong Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Hang Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Weibing Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Hua Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
9
|
Dimou A, Nasarre C, Peterson YK, Pagano R, Gooz M, Nasarre P, Drabkin HA, Armeson KE, Gibney BC, Gemmill RM, Denlinger CE. Neuropilin-2b facilitates resistance to tyrosine kinase inhibitors in non-small cell lung cancer. J Thorac Cardiovasc Surg 2020; 162:463-473. [PMID: 32653291 DOI: 10.1016/j.jtcvs.2020.03.166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Innate and acquired resistance is the principle factor limiting the efficacy of tyrosine kinase inhibitors in lung cancer. We have observed a dramatic upregulation of the cell surface co-receptor neuropilin-2b in lung cancers clinically treated with tyrosine kinase inhibitors correlating with acquired resistance. We hypothesize that neuropilin-2b plays a functional role in acquired tyrosine kinase inhibitor resistance. METHODS Non-small cell lung cancer proliferation and survival were determined during chronic tyrosine kinase inhibitor exposure in the presence or absence of neuropilin-2b knock-down. Interactions of neuropilin-2a and neuropilin-2b isoforms with PTEN and GSK3β were assessed by immunoprecipitation. Neuropilin-2a and neuropilin-2b mutants deleted for their cytoplasmic domains were used to identify regions responsible for neuropilin-2b-GSK3β interaction. Because GSK3β is known to phosphorylate and degrade PTEN, phospho-PTEN and total PTEN levels were assessed after transfection of neuropilin-2a and neuropilin-2b wild-type and mutant constructs. RESULTS Non-small cell lung cancer cells chronically treated with gefitinib or osimertinib developed drug resistance and exhibited logarithmic growth in the presence of endothelial growth factor receptor tyrosine kinase inhibitors. However, neuropilin-2b knockdown cells remained sensitive to gefitinib. Likewise, neuropilin-2b knockdown suppressed and neuropilin-2a knockdown enhanced cellular migration. Acquired drug resistance and cell migration correlated with neuropilin-2b-dependent AKT activation with the intermediate step of GSK3β-dependent PTEN degradation. A specific binding site for GSK3β on the cytoplasmic domain of neuropilin-2b was identified with truncated protein constructs and computer modeling. CONCLUSIONS Neuropilin-2b facilitates non-small cell lung cancer resistance to tyrosine kinase inhibitors, and this biological effect relates to AKT activation. Neuropilin-2b GSK3β interactions appear to be essential for PTEN degradation and AKT activation in lung cancer cells. Disruption of the neuropilin-2b GSK3β interaction may represent a novel treatment strategy to preserve sensitivity to tyrosine kinase inhibitors in non-small cell lung cancer.
Collapse
Affiliation(s)
- Anastasios Dimou
- Division of Medical Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Cecile Nasarre
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC
| | - Rose Pagano
- Division of Medical Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Monika Gooz
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC
| | - Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Harry A Drabkin
- Division of Medical Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Kent E Armeson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Barry C Gibney
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Robert M Gemmill
- Division of Medical Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Chadrick E Denlinger
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC.
| |
Collapse
|
10
|
Kumaravel S, Singh S, Roy S, Venkatasamy L, White TK, Sinha S, Glaser SS, Safe SH, Chakraborty S. CXCL11-CXCR3 Axis Mediates Tumor Lymphatic Cross Talk and Inflammation-Induced Tumor, Promoting Pathways in Head and Neck Cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:900-915. [PMID: 32035061 PMCID: PMC7180517 DOI: 10.1016/j.ajpath.2019.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Tumor metastasis to the draining lymph nodes is critical in patient prognosis and is tightly regulated by molecular interactions mediated by lymphatic endothelial cells (LECs). The underlying mechanisms remain undefined in the head and neck squamous cell carcinomas (HNSCCs). Using HNSCC cells and LECs we determined the mechanisms mediating tumor-lymphatic cross talk. The effects of a pentacyclic triterpenoid, methyl 2-trifluoromethyl-3,11-dioxoolean-1,12-dien-30-oate (CF3DODA-Me), a potent anticancer agent, were studied on cancer-lymphatic interactions. In response to inflammation, LECs induced the chemokine (C-X-C motif) ligand 9/10/11 chemokines with a concomitant increase in the chemokine (C-X-C motif) receptor 3 (CXCR3) in tumor cells. CF3DODA-Me showed antiproliferative effects on tumor cells, altered cellular bioenergetics, suppressed matrix metalloproteinases and chemokine receptors, and the induction of CXCL11-CXCR3 axis and phosphatidylinositol 3-kinase/AKT pathways. Tumor cell migration to LECs was inhibited by blocking CXCL11 whereas recombinant CXCL11 significantly induced tumor migration, epithelial-to-mesenchymal transition, and matrix remodeling. Immunohistochemical analysis of HNSCC tumor arrays showed enhanced expression of CXCR3 and increased lymphatic vessel infiltration. Furthermore, The Cancer Genome Atlas RNA-sequencing data from HNSCC patients also showed a positive correlation between CXCR3 expression and lymphovascular invasion. Collectively, our data suggest a novel mechanism for cross talk between the LECs and HNSCC tumors through the CXCR3-CXCL11 axis and elucidate the role of the triterpenoid CF3DODA-Me in abrogating several of these tumor-promoting pathways.
Collapse
Affiliation(s)
- Subhashree Kumaravel
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Sumeet Singh
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Sukanya Roy
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Lavanya Venkatasamy
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Tori K White
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Samiran Sinha
- Department of Statistics, Texas A&M University, College Station, Texas
| | - Shannon S Glaser
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas
| | - Stephen H Safe
- Department Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, Texas.
| |
Collapse
|
11
|
Wang C, Liu Q, Huang M, Zhou Q, Zhang X, Zhang J, Xie R, Yu Y, Chen S, Fan J, Chen X. Loss of GATA6 expression promotes lymphatic metastasis in bladder cancer. FASEB J 2020; 34:5754-5766. [PMID: 32103545 DOI: 10.1096/fj.201903176r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 12/19/2022]
Abstract
Lymph node metastasis is associated with tumor relapse and poor patient prognosis in bladder cancer. However, the mechanisms by which bladder carcinoma cells induce lymphangiogenesis and further promote metastasis in the lymphatic system remain unclear. Here, we show that the transcription factor GATA-binding factor 6 (GATA6) was substantially downregulated in bladder cancer via promoter hypermethylation. Low-level GATA6 expression significantly correlated with lymph node metastasis positivity and was able to predict earlier relapse and shorter survival of bladder cancer. Reconstitution of GATA6 inhibited lymphangiogenesis and lymph node metastasis in GATA6-low bladder cancer cells, while silencing of GATA6 rendered lymphatic metastasis in GATA6-high bladder cancer cells. Additionally, we demonstrated that GATA6 bound to the promoter of vascular endothelial growth factor (VEGF)-C, a lymphangiogenic factor, and acted as a transcriptional repressor. This GATA6/VEGF-C axis was essential for GATA6-mediated lymphatic metastasis. In bladder cancer patients, low GATA6 correlated with high VEGF-C and reduced overall survival. These findings indicate GATA6 as a pivotal regulator in the lymphatic dissemination of bladder cancer and suggest a new therapeutic target for the disease.
Collapse
Affiliation(s)
- Chanjuan Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qinghua Liu
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ming Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingtong Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruihui Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanqi Yu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shang Chen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianbing Fan
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
12
|
Wu ZS, Ding W, Cai J, Bashir G, Li YQ, Wu S. Communication Of Cancer Cells And Lymphatic Vessels In Cancer: Focus On Bladder Cancer. Onco Targets Ther 2019; 12:8161-8177. [PMID: 31632067 PMCID: PMC6781639 DOI: 10.2147/ott.s219111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/07/2019] [Indexed: 12/16/2022] Open
Abstract
Bladder cancer is one of the most commonly diagnosed cancers worldwide and causes the highest lifetime treatment costs per patient. Bladder cancer is most likely to metastasize through lymphatic ducts, and once the lymph nodes are involved, the prognosis is poorly and finitely improved by current modalities. The underlying metastatic mechanism for bladder cancer is thus becoming a research focus to date. To identify relevant published data, an online search of the PubMed/Medline archives was performed to locate original articles and review articles regarding lymphangiogenesis and lymphatic metastasis in urinary bladder cancer (UBC), and was limited to articles in English published between 1998 and 2018. A further search of the clinical trials.gov search engine was conducted to identify both trials with results available and those with results not yet available. Herein, we summarized the unique mechanisms and biomarkers involved in the malignant progression of bladder cancer as well as their emerging roles in therapeutics, and that current data suggests that lymphangiogenesis and lymph node invasion are important prognostic factors for UBC. The growing knowledge about their roles in bladder cancers provides the basis for novel therapeutic strategies. In addition, more basic and clinical research needs to be conducted in order to identify further accurate predictive molecules and relevant mechanisms.
Collapse
Affiliation(s)
- Zhang-song Wu
- Medical College, Shenzhen University, Shenzhen518000, People’s Republic of China
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Wa Ding
- Medical College, Shenzhen University, Shenzhen518000, People’s Republic of China
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Jiajia Cai
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
- Medical College, Anhui University of Science and Technology, Huainan232001, People’s Republic of China
| | - Ghassan Bashir
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Yu-qing Li
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Song Wu
- Medical College, Shenzhen University, Shenzhen518000, People’s Republic of China
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
- Shenzhen following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen518000, People’s Republic of China
- Medical College, Anhui University of Science and Technology, Huainan232001, People’s Republic of China
| |
Collapse
|
13
|
Kataru RP, Ly CL, Shin J, Park HJ, Baik JE, Rehal S, Ortega S, Lyden D, Mehrara BJ. Tumor Lymphatic Function Regulates Tumor Inflammatory and Immunosuppressive Microenvironments. Cancer Immunol Res 2019; 7:1345-1358. [PMID: 31186247 DOI: 10.1158/2326-6066.cir-18-0337] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/17/2018] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Proliferation of aberrant, dysfunctional lymphatic vessels around solid tumors is a common histologic finding. Studies have shown that abnormalities in lymphatic function result in accumulation of inflammatory cells with an immunosuppressive profile. We tested the hypothesis that dysfunctional lymphatic vessels surrounding solid tumors regulate changes in the tumor microenvironment and tumor-specific immune responses. Using subcutaneously implanted mouse melanoma and breast cancer tumors in a lymphatic endothelial cell-specific diphtheria toxin receptor transgenic mouse, we found that local ablation of lymphatic vessels increased peritumoral edema, as compared with controls. Comparative analysis of the peritumoral fluid demonstrated increases in the number of macrophages, CD4+ inflammatory cells, F4/80+/Gr-1+ (myeloid-derived suppressor cells), CD4+/Foxp3+ (Tregs) immunosuppressive cells, and expression of inflammatory cytokines such as TNFα, IFNγ, and IL1β following lymphatic ablation. Tumors grown in lymphatic ablated mice exhibited reduced intratumoral accumulation of cytotoxic T cells and increased tumor PD-L1 expression, causing rapid tumor growth, compared with tumors grown in nonlymphatic-ablated mice. Our study suggests that lymphatic dysfunction plays a role in regulating tumor microenvironments and may be therapeutically targeted in combination with immunotherapy to prevent tumor growth and progression.
Collapse
Affiliation(s)
- Raghu P Kataru
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Catherine L Ly
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jinyeon Shin
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hyeung Ju Park
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jung Eun Baik
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sonia Rehal
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sagrario Ortega
- Transgenic Mice Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Babak J Mehrara
- Department of Surgery, Plastic and Reconstructive Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
14
|
Chang CW, Seibel AJ, Song JW. Application of microscale culture technologies for studying lymphatic vessel biology. Microcirculation 2019; 26:e12547. [PMID: 30946511 DOI: 10.1111/micc.12547] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/04/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022]
Abstract
Immense progress in microscale engineering technologies has significantly expanded the capabilities of in vitro cell culture systems for reconstituting physiological microenvironments that are mediated by biomolecular gradients, fluid transport, and mechanical forces. Here, we examine the innovative approaches based on microfabricated vessels for studying lymphatic biology. To help understand the necessary design requirements for microfluidic models, we first summarize lymphatic vessel structure and function. Next, we provide an overview of the molecular and biomechanical mediators of lymphatic vessel function. Then we discuss the past achievements and new opportunities for microfluidic culture models to a broad range of applications pertaining to lymphatic vessel physiology. We emphasize the unique attributes of microfluidic systems that enable the recapitulation of multiple physicochemical cues in vitro for studying lymphatic pathophysiology. Current challenges and future outlooks of microscale technology for studying lymphatics are also discussed. Collectively, we make the assertion that further progress in the development of microscale models will continue to enrich our mechanistic understanding of lymphatic biology and physiology to help realize the promise of the lymphatic vasculature as a therapeutic target for a broad spectrum of diseases.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Alex J Seibel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio.,The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
15
|
In vivo near infrared fluorescence imaging and dynamic quantification of pancreatic metastatic tumors using folic acid conjugated biodegradable mesoporous silica nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1867-1877. [PMID: 29733890 DOI: 10.1016/j.nano.2018.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 11/21/2022]
Abstract
Cancer metastasis is one of the biggest challenges in cancer treatments since it increases the likelihood that a patient will die from the disease. Therefore, the availability of techniques for the early detection and quantification of tumors is very important. We have prepared cyanine 7.5 NHS ester (Cy7.5) and folic acid (FA) conjugated biodegradable mesoporous silica nanoparticles (bMSN@Cy7.5-FA NPs) (~100 nm) for visualizing tumors in vivo. The fluorescence spectra revealed that the emission peak of bMSN@Cy7.5-FA NPs had a red-shift of 1 nm. Confocal immunofluorescent images showed that bMSN@Cy7.5-FA NPs had an excellent targeting ability for visualizing cancer cells. In vivo fluorescence imaging has been conducted using an orthotopic model for pancreatic cancer within 48 h, and the fluorescence intensity reached a maximum at a post injection time-point of 12 h, which demonstrated that the use of bMSN@Cy7.5-FA NPs provides an excellent imaging platform for tumor precision therapy in mice.
Collapse
|
16
|
Parmar P, Marwah N, Parshad S, Yadav T, Batra A, Sen R. Clinicopathological Significance of Tumor Lymphatic Vessel Density in Head and Neck Squamous Cell Carcinoma. Indian J Otolaryngol Head Neck Surg 2018; 70:102-110. [PMID: 29456952 DOI: 10.1007/s12070-017-1216-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/03/2017] [Indexed: 01/10/2023] Open
Abstract
Various studies have demonstrated that the lymphatic system is the additional route for solid tumor metastasis. Lymph nodes metastasis in Head and neck squamous cell carcinoma (HNSCC) is a major prognostic indicator for disease progression and a guide for therapeutic strategies. We conducted a study to compare intratumoral (IT) and peritumoral (PT) lymphatic vessel density (LVD) in HNSCC using lymphatic marker D2-40 and its correlation with lymph node metastasis, histological grading and other clinicopathological parameters. Fifty specimen of HNSCC with modified radical neck dissection tissue were included in the study group. Tissue from tumor, peritumoral tissue, tumor margin and all the lymph nodes were processed for paraffin wax blocks and histopathological diagnosis. Immunohistochemical profile of lymphatic vessels in intratumoral and peritumoral tissue was assessed by subjecting one section each from the tumor and peritumoral tissue to D2-40 immunostain. To determine LVD, four fields with the highest LVD (hot spots) were identified. The mean values were calculated by taking an average of all the measurements. The comparison of LVD between peritumoral and intratumoral area revealed significantly higher PT-LVD (P = 0.001). No significant association was seen between LVD, IT-LVD and PT-LVD and different age groups, gender, site of tumor, risk factors, size of tumor, tumor inflammation, pushing/infiltrating margin and stage of tumors. Significantly higher LVD, IT-LVD and PT-LVD was seen in association with lymph node metastasis. Both high intratumoral and peritumoral LVD were found significantly associated with the presence of lymph node metastasis, however lymphatic vessels were found to be significantly more numerous and larger in peritumoral areas as compared to intratumoral lymphatics. The specificity of D2-40 as a lymphatic endothelial marker was also confirmed. The results of our study support the possibility of using the determination of tumor lymphangiogenesis to identify patients of HNSCC who are at risk of developing the lymph node metastasis.
Collapse
Affiliation(s)
- Padam Parmar
- 1Department of Pathology, PT BDS PGIMS, Rohtak, Haryana 124001 India
| | - Nisha Marwah
- 1Department of Pathology, PT BDS PGIMS, Rohtak, Haryana 124001 India
| | | | - Taruna Yadav
- 3Department of Radiodiagnosis, PT BDS PGIMS, Rohtak, Haryana India
| | - Ashima Batra
- 1Department of Pathology, PT BDS PGIMS, Rohtak, Haryana 124001 India
| | - Rajeev Sen
- 1Department of Pathology, PT BDS PGIMS, Rohtak, Haryana 124001 India
| |
Collapse
|
17
|
Zhan Y, Zhan W, Li H, Xu X, Cao X, Zhu S, Liang J, Chen X. In Vivo Dual-Modality Fluorescence and Magnetic Resonance Imaging-Guided Lymph Node Mapping with Good Biocompatibility Manganese Oxide Nanoparticles. Molecules 2017; 22:molecules22122208. [PMID: 29231865 PMCID: PMC6149721 DOI: 10.3390/molecules22122208] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 01/19/2023] Open
Abstract
Multifunctional manganese oxide nanoparticles (NPs) with impressive enhanced T₁ contrast ability show great promise in biomedical diagnosis. Herein, we developed a dual-modality imaging agent system based on polyethylene glycol (PEG)-coated manganese oxide NPs conjugated with organic dye (Cy7.5), which functions as a fluorescence imaging (FI) agent as well as a magnetic resonance imaging (MRI) imaging agent. The formed Mn₃O₄@PEG-Cy7.5 NPs with the size of ~10 nm exhibit good colloidal stability in different physiological media. Serial FI and MRI studies that non-invasively assessed the bio-distribution pattern and the feasibility for in vivo dual-modality imaging-guided lymph node mapping have been investigated. In addition, histological and biochemical analyses exhibited low toxicity even at a dose of 20 mg/kg in vivo. Since Mn₃O₄@PEG-Cy7.5 NPs exhibited desirable properties as imaging agents and good biocompatibility, this work offers a robust, safe, and accurate diagnostic platform based on manganese oxide NPs for tumor metastasis diagnosis.
Collapse
Affiliation(s)
- Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
- Xidian-Ningbo Information Technology Institute, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Wenhua Zhan
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Hanrui Li
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Xinyi Xu
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Xu Cao
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Shouping Zhu
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, Shaanxi, China.
| |
Collapse
|
18
|
Ong HS, Gokavarapu S, Xu Q, Tian Z, Li J, Ji T, Zhang CP. Cytoplasmic neuropilin 2 is associated with metastasis and a poor prognosis in early tongue cancer patients. Int J Oral Maxillofac Surg 2017; 46:1205-1219. [PMID: 28602571 DOI: 10.1016/j.ijom.2017.03.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 03/17/2017] [Accepted: 03/28/2017] [Indexed: 11/27/2022]
Abstract
Neuropilin 2 (Nrp2) plays an important role in regulating lymphangiogenesis. Nrp2 expression in early tongue cancer was investigated to predict lymph node metastasis and the long-term prognosis. The relationships between clinicopathological variables of cT1-T2N0 tongue squamous cell carcinoma (SCC) and overexpression of Nrp2, vascular endothelial growth factor C (VEGFC), vascular endothelial growth factor receptor 3 (VEGFR3), and semaphorin 3F (Sema3F) were analyzed. Expression levels were compared using oral SCC cell lines. The Nrp2 gene was silenced to determine the impact of Nrp2. Cytoplasmic Nrp2 overexpression predicted regional metastasis with sensitivity and specificity of 90.3% and 42.1%, respectively. Cytoplasmic Nrp2 overexpression (P<0.001) and VEGFC overexpression (P=0.006) were significantly related to regional metastasis (Student t-test). However, only cytoplasmic Nrp2 overexpression was an independent prognostic factor for both disease-free survival (DFS; P=0.008) and overall survival (OS; P=0.016) (Cox regression); the risk of recurrence was 12-times higher (P=0.015) and risk of mortality was 8-times higher (P=0.016). Co-localization of Nrp2 and VEGFC was greater within the cytoplasm of aggressive cell lines (HN12 and RCa-T). Nrp2 plays a role in tumourigenesis; VEGFC supplementation cannot rescue the biological function of Nrp2 in Nrp2-depleted cell lines. Cytoplasmic Nrp2 overexpression is associated with decreased OS and DFS. Cytoplasmic Nrp2 overexpression may be a reliable diagnostic and prognostic marker for early tongue SCC.
Collapse
Affiliation(s)
- H S Ong
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - S Gokavarapu
- Head and Neck Oncology Reconstructive Surgery, Department of Surgical Oncology, Krishna Institute of Medical Science, Hyderabad, Telangana, India
| | - Q Xu
- Shanghai Key Laboratory of Oral and Maxillofacial-Head and Neck Oncology and Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Z Tian
- Department of Oral Pathology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - J Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - T Ji
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - C P Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Paquet-Fifield S, Roufail S, Zhang YF, Sofian T, Byrne DJ, Coughlin PB, Fox SB, Stacker SA, Achen MG. The fibrinolysis inhibitor α 2-antiplasmin restricts lymphatic remodelling and metastasis in a mouse model of cancer. Growth Factors 2017; 35:61-75. [PMID: 28697634 DOI: 10.1080/08977194.2017.1349765] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Remodelling of lymphatic vessels in tumours facilitates metastasis to lymph nodes. The growth factors VEGF-C and VEGF-D are well known inducers of lymphatic remodelling and metastasis in cancer. They are initially produced as full-length proteins requiring proteolytic processing in order to bind VEGF receptors with high affinity and thereby promote lymphatic remodelling. The fibrinolytic protease plasmin promotes processing of VEGF-C and VEGF-D in vitro, but its role in processing them in cancer was unknown. Here we explore plasmin's role in proteolytically activating VEGF-D in vivo, and promoting lymphatic remodelling and metastasis in cancer, by co-expressing the plasmin inhibitor α2-antiplasmin with VEGF-D in a mouse tumour model. We show that α2-antiplasmin restricts activation of VEGF-D, enlargement of intra-tumoural lymphatics and occurrence of lymph node metastasis. Our findings indicate that the fibrinolytic system influences lymphatic remodelling in tumours which is consistent with previous clinicopathological observations correlating fibrinolytic components with cancer metastasis.
Collapse
Affiliation(s)
- Sophie Paquet-Fifield
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Sally Roufail
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - You-Fang Zhang
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Trifina Sofian
- b Australian Centre for Blood Diseases , Monash University , Prahran, Melbourne , Australia
| | - David J Byrne
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- c Department of Pathology , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Paul B Coughlin
- b Australian Centre for Blood Diseases , Monash University , Prahran, Melbourne , Australia
- d Eastern Health , Box Hill , Australia
| | - Stephen B Fox
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- c Department of Pathology , Peter MacCallum Cancer Centre , Melbourne , Australia
- e Sir Peter MacCallum Department of Oncology , University of Melbourne , Parkville , Australia
| | - Steven A Stacker
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- e Sir Peter MacCallum Department of Oncology , University of Melbourne , Parkville , Australia
| | - Marc G Achen
- a Tumour Angiogenesis and Microenvironment Program , Peter MacCallum Cancer Centre , Melbourne , Australia
- e Sir Peter MacCallum Department of Oncology , University of Melbourne , Parkville , Australia
| |
Collapse
|
20
|
Maisel K, Sasso MS, Potin L, Swartz MA. Exploiting lymphatic vessels for immunomodulation: Rationale, opportunities, and challenges. Adv Drug Deliv Rev 2017; 114:43-59. [PMID: 28694027 PMCID: PMC6026542 DOI: 10.1016/j.addr.2017.07.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Lymphatic vessels are the primary route of communication from peripheral tissues to the immune system; as such, they represent an important component of local immunity. In addition to their transport functions, new immunomodulatory roles for lymphatic vessels and lymphatic endothelial cells have come to light in recent years, demonstrating that lymphatic vessels help shape immune responses in a variety of ways: promoting tolerance to self-antigens, archiving antigen for later presentation, dampening effector immune responses, and resolving inflammation, among others. In addition to these new biological insights, the growing field of immunoengineering has begun to explore therapeutic approaches to utilize or exploit the lymphatic system for immunotherapy.
Collapse
Affiliation(s)
- Katharina Maisel
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Maria Stella Sasso
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Lambert Potin
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Melody A Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; Ben May Institute for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Abstract
Lymphatic metastasis is an important event in the progress of metastasis in colorectal cancer (CRC). The purpose of this article is to assess the role of lymphangiogenesis on CRC. In peritumoral areas of CRC, the lymphatic microvessel density (LMVD) is higher than those in normal colorectal tissues. Morever, the high LMVD is correlated with DFS and local recurrence in CRC. The VEGF-C/VEGF-D/VEGFR-3 pathway, sonic hedgehog (Shh) signaling pathway and extracellular matrix (ECM) are involved in the regulation of lymphangiogenesis in CRC. Inhibition of the VEGF-C/VEGF-D/VEGFR-3 pathway by specific antibodies has been reported to efficiently inhibit experimental tumor lymphangiogenesis and metastasis in animal experiments. Although lymphangiogenesis has been reported to play an important role in the occurrence of colon cancer and to be associated with prognosis, it remains unclear whether it is a valid therapeutic target molecule. Further study of the potential of targeting this process for anti-lymphatic therapies is worthwhile.
Collapse
Affiliation(s)
- Ciyou Huang
- Department of Endocrinology, Wuxi Second Hospital, Nanjing Medical University, Jiangsu, China. E-mail.
| | | |
Collapse
|
22
|
Hendry SA, Farnsworth RH, Solomon B, Achen MG, Stacker SA, Fox SB. The Role of the Tumor Vasculature in the Host Immune Response: Implications for Therapeutic Strategies Targeting the Tumor Microenvironment. Front Immunol 2016; 7:621. [PMID: 28066431 PMCID: PMC5168440 DOI: 10.3389/fimmu.2016.00621] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022] Open
Abstract
Recently developed cancer immunotherapy approaches including immune checkpoint inhibitors and chimeric antigen receptor T cell transfer are showing promising results both in trials and in clinical practice. These approaches reflect increasing recognition of the crucial role of the tumor microenvironment in cancer development and progression. Cancer cells do not act alone, but develop a complex relationship with the environment in which they reside. The host immune response to tumors is critical to the success of immunotherapy; however, the determinants of this response are incompletely understood. The immune cell infiltrate in tumors varies widely in density, composition, and clinical significance. The tumor vasculature is a key component of the microenvironment that can influence tumor behavior and treatment response and can be targeted through the use of antiangiogenic drugs. Blood vascular and lymphatic endothelial cells have important roles in the trafficking of immune cells, controlling the microenvironment, and modulating the immune response. Improving access to the tumor through vascular alteration with antiangiogenic drugs may prove an effective combinatorial strategy with immunotherapy approaches and might be applicable to many tumor types. In this review, we briefly discuss the host's immune response to cancer and the treatment strategies utilizing this response, before focusing on the pathological features of tumor blood and lymphatic vessels and the contribution these might make to tumor immune evasion.
Collapse
Affiliation(s)
- Shona A Hendry
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Rae H Farnsworth
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Benjamin Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Marc G Achen
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia; Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Steven A Stacker
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia; Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| |
Collapse
|
23
|
Abstract
BACKGROUND Metastasis is the main cause of mortality in cancer patients. Two major routes of cancer cell spread are currently being recognized: dissemination via blood vessels (hematogenous spread) and dissemination via the lymphatic system (lymphogenous spread). Here, our current knowledge on the role of both blood and lymphatic vessels in cancer cell metastasis is summarized. In addition, I will discuss why cancer cells select one or both of the two routes to disseminate and I will provide a short description of the passive and active models of intravasation. Finally, lymphatic vessel density (LVD), blood vessel density (BVD), interstitial fluid pressure (IFP) and tumor hypoxia, as well as regional lymph node metastasis and the recently discovered primo vascular system (PVS) will be highlighted as important factors influencing tumor cell motility and spread and, ultimately, clinical outcome. CONCLUSIONS Lymphangiogenesis and angiogenesis are important phenomena involved in the spread of cancer cells and they are associated with a poor prognosis. It is anticipated that new discoveries and advancing knowledge on these phenomena will allow an improvement in the treatment of cancer patients.
Collapse
Affiliation(s)
- Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
- Department of General Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079, Lublin, Poland.
| |
Collapse
|
24
|
Phase 1 study of the anti-vascular endothelial growth factor receptor 3 monoclonal antibody LY3022856/IMC-3C5 in patients with advanced and refractory solid tumors and advanced colorectal cancer. Cancer Chemother Pharmacol 2016; 78:815-24. [PMID: 27566701 DOI: 10.1007/s00280-016-3134-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/11/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Metastasis of solid tumors to regional lymph nodes is facilitated by tumor lymphangiogenesis, which is primarily mediated by the vascular endothelial growth factor receptor 3 (VEGFR-3). We conducted a phase 1 dose-escalation (part A) study of the VEGFR-3 human immunoglobulin G subclass 1 monoclonal antibody LY3022856 in advanced solid tumors, followed by a colorectal cancer (CRC) expansion (part B). METHODS Part A evaluated the safety profile and maximum tolerated dose (MTD) of LY3022856 in patients treated intravenously at doses of 5-30 mg/kg weekly (qwk). Part B further evaluated tolerability in CRC patients treated with 30 mg/kg. Secondary objectives were pharmacokinetics, anti-tumor activity, and pharmacodynamics (exploratory). RESULTS A total of 44 patients (23 in part A; 21 in part B) were treated; only one dose-limiting toxicity was observed at the lowest dose level. The MTD was not reached. Treatment-emergent adverse events (TEAEs) of any grade included in ≥15 % of all patients were: nausea (41 %), fatigue (32 %), vomiting (30 %), decreased appetite (27 %), pyrexia (25 %), peripheral edema (23 %), and urinary tract infection (UTI, 20 %). The most common grade 3/4 TEAEs included UTI and small intestinal obstruction (7 % each). No radiographic responses were noted. Median progression-free survival in part B was 6.3 weeks (95 % confidence interval: 5.1, 14.4), and a best overall response of stable disease was observed in 4 CRC patients (19.0 %). CONCLUSIONS LY3022856 was well tolerated up to a dose of 30 mg/kg qwk, but with minimal anti-tumor activity in CRC. CLINICALTRIALS. GOV IDENTIFIER NCT01288989.
Collapse
|
25
|
Teng H, Yang Y, Wei H, Liu Z, Liu Z, Ma Y, Gao Z, Hou L, Zou X. Fucoidan Suppresses Hypoxia-Induced Lymphangiogenesis and Lymphatic Metastasis in Mouse Hepatocarcinoma. Mar Drugs 2015; 13:3514-30. [PMID: 26047481 PMCID: PMC4483642 DOI: 10.3390/md13063514] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/19/2015] [Indexed: 12/30/2022] Open
Abstract
Metastasis, the greatest clinical challenge associated with cancer, is closely connected to multiple biological processes, including invasion and adhesion. The hypoxic environment in tumors is an important factor that causes tumor metastasis by activating HIF-1α. Fucoidan, extracted from brown algae, is a sulfated polysaccharide and, as a novel marine biological material, has been used to treat various disorders in China, Korea, Japan and other countries. In the present study, we demonstrated that fucoidan derived from Undaria pinnatifida sporophylls significantly inhibits the hypoxia-induced expression, nuclear translocation and activity of HIF-1α, the synthesis and secretion of VEGF-C and HGF, cell invasion and lymphatic metastasis in a mouse hepatocarcinoma Hca-F cell line. Fucoidan also suppressed lymphangiogenesis in vitro and in vivo. In addition, accompanied by a reduction in the HIF-1α nuclear translocation and activity, fucoidan significantly reduced the levels of p-PI3K, p-Akt, p-mTOR, p-ERK, NF-κB, MMP-2 and MMP-9, but increased TIMP-1 levels. These results indicate strongly that the anti-metastasis and anti-lymphangiogenesis activities of fucoidan are mediated by suppressing HIF-1α/VEGF-C, which attenuates the PI3K/Akt/mTOR signaling pathways.
Collapse
Affiliation(s)
- Hongming Teng
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Yazong Yang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Hengyun Wei
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Zundong Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Zhichao Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Yanhong Ma
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Zixiang Gao
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| | - Lin Hou
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
| | - Xiangyang Zou
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China; E-Mails: (H.T.); yangyazong123.@126.com (Y.Y.); (H.W.); (Z.L.); (Z.L.); (Y.M.); (Z.G.)
| |
Collapse
|
26
|
Chang YW, Su CM, Su YH, Ho YS, Lai HH, Chen HA, Kuo ML, Hung WC, Chen YW, Wu CH, Chen PS, Su JL. Novel peptides suppress VEGFR-3 activity and antagonize VEGFR-3-mediated oncogenic effects. Oncotarget 2015; 5:3823-35. [PMID: 25003617 PMCID: PMC4116523 DOI: 10.18632/oncotarget.1709] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Vascular endothelial growth factor receptor 3 (VEGFR-3) supports tumor lymphangiogenesis. It was originally identified as a lymphangiogenic factor expressed in lymphatic endothelial cells. VEGFR-3 was detected in advanced human malignancies and correlated with poor prognosis. Our previous studies show that activation of the VEGF-C/VEGFR-3 axis promotes cancer metastasis and is associated with clinical progression in patients with lung cancer, indicating that VEGFR-3 is a potential target for cancer therapy. In this study, we developed eight peptides targeting VEGFR-3. Two peptides strongly inhibited the kinase activity of VEGFR-3 and suppressed VEGF-C-mediated invasion of cancer cells. Moreover, these peptides abolished VEGF-C-induced drug resistance and tumor initiating cell formation. This study demonstrates the therapeutic potential of VEGFR-3-targeting peptides.
Collapse
Affiliation(s)
- Yi-Wen Chang
- Graduate Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan; Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | | - Chih-Hsiung Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University
| | - Pai-Sheng Chen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan; Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Jen-Liang Su
- National Institute of Cancer Research, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Cancer Biology, College of Medicine, China Medical University, Taichung 404, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 404, Taiwan
| |
Collapse
|
27
|
Shang L, Zhao J, Wang W, Xiao W, Li J, Li X, Song W, Liu J, Wen F, Yue C. [Inhibitory effect of endostar on lymphangiogenesis in non-small cell lung cancer and its effect on circulating tumor cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 17:722-9. [PMID: 25342038 PMCID: PMC6000404 DOI: 10.3779/j.issn.1009-3419.2014.10.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
背景与目的 血管内皮抑素可以抑制肿瘤新生血管的生成,但对肿瘤微淋巴管的形成与发展是否存在抑制效应引起我们关注。本研究旨在探讨重组人血管内皮抑素(recombinant human endostatin injection)对非小细胞肺癌组织中血管内皮生长因子(vascular endothelial growth factor, VEGF)-C、VEGF-D和VEGF受体(VEGFR)-3表达及对外周血循环肿瘤细胞数目的影响。 方法 荷瘤裸鼠随机分为空白对照组、顺铂组、不同浓度重组人血管内皮抑素组及重组人血管内皮抑素+顺铂组,连续给药2周。1周后检测肿瘤组织中VEGF-C、VEGF-D、VEGFR-3的表达水平和微淋巴管密度。免疫荧光染色诊断和计数循环肿瘤细胞。 结果 重组人血管内皮抑素组与重组人血管内皮抑素联合顺铂组的表达阳性率、微淋巴管密度均明显低于空白对照组与顺铂组(P < 0.05);较高浓度的重组人血管内皮抑素联合顺铂组与重组人血管内皮抑素组表达阳性率和微淋巴管密度低于相应较低重组人血管内皮抑素浓度的组别(P < 0.05)。各组微淋巴管密度与VEGF-C、VEGF-D、VEGFR-3表达阳性率存在正相关。重组人血管内皮抑素联合顺铂各组的循环肿瘤细胞数目明显低于单独使用顺铂或重组人血管内皮抑素(P < 0.05)。 结论 重组人血管内皮抑素可以抑制肿瘤新生淋巴管生成,减少循环肿瘤细胞,作用大小与浓度有关。与顺铂联合使用能够更有效的减少循环肿瘤细胞。
Collapse
Affiliation(s)
- Liqun Shang
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Jie Zhao
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Wei Wang
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Wang Xiao
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Jun Li
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Xuechang Li
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Weian Song
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Junqiang Liu
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Feng Wen
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| | - Caiying Yue
- Department of Thoracic Surgery, PLA Navy General Hospital, Beijing 100048, China
| |
Collapse
|
28
|
Yang Y, Chen XH, Li FG, Chen YX, Gu LQ, Zhu JK, Li P. In VitroInduction of Human Adipose-Derived Stem Cells into Lymphatic Endothelial-Like Cells. Cell Reprogram 2015; 17:69-76. [PMID: 25647247 DOI: 10.1089/cell.2014.0043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Yi Yang
- Department of Microsurgery and Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- These authors contributed equally to this work
| | - Xiao-hu Chen
- Department of Orthopedic Trauma, The Hui Ya Hospital of Sun Yat-sen University, Huizhou, 516000, China
- These authors contributed equally to this work
| | - Fu-gui Li
- Department of Cancer Institute, The Zhong Shan Hospital of Sun Yat-sen University, Zhongshan, 528403, China
| | - Yun-xian Chen
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-qiang Gu
- Department of Microsurgery and Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-kai Zhu
- Department of Microsurgery and Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ping Li
- Department of Microsurgery and Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
29
|
Lee E, Pandey NB, Popel AS. Crosstalk between cancer cells and blood endothelial and lymphatic endothelial cells in tumour and organ microenvironment. Expert Rev Mol Med 2015; 17:e3. [PMID: 25634527 PMCID: PMC4352000 DOI: 10.1017/erm.2015.2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tumour and organ microenvironments are crucial for cancer progression and metastasis. Crosstalk between multiple non-malignant cell types in the microenvironments and cancer cells promotes tumour growth and metastasis. Blood and lymphatic endothelial cells (BEC and LEC) are two of the components in the microenvironments. Tumour blood vessels (BV), comprising BEC, serve as conduits for blood supply into the tumour, and are important for tumour growth as well as haematogenous tumour dissemination. Lymphatic vessels (LV), comprising LEC, which are relatively leaky compared with BV, are essential for lymphogenous tumour dissemination. In addition to describing the conventional roles of the BV and LV, we also discuss newly emerging roles of these endothelial cells: their crosstalk with cancer cells via molecules secreted by the BEC and LEC (also called angiocrine and lymphangiocrine factors). This review suggests that BEC and LEC in various microenvironments can be orchestrators of tumour progression and proposes new mechanism-based strategies to discover new therapies to supplement conventional anti-angiogenic and anti-lymphangiogenic therapies.
Collapse
Affiliation(s)
- Esak Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Niranjan B. Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
30
|
Alyahya R, Sudha T, Racz M, Stain SC, Mousa SA. Anti-metastasis efficacy and safety of non-anticoagulant heparin derivative versus low molecular weight heparin in surgical pancreatic cancer models. Int J Oncol 2014; 46:1225-31. [PMID: 25530018 DOI: 10.3892/ijo.2014.2803] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/27/2014] [Indexed: 11/06/2022] Open
Abstract
Heparin and its derivatives are known to attenuate cancer metastasis in preclinical models, but have not been used clinically due to adverse bleeding effects. This study compared the efficacy of S-NACH (a sulfated non-anticoagulant heparin) versus tinzaparin (a low molecular weight heparin) in inhibiting metastasis of a growing primary tumor and following surgical excision of primary tumor in a pancreatic cancer mouse model. The efficacy of S-NACH versus tinzaparin on metastasis of the primary tumor was evaluated in each experiment using IVIS imaging. Athymic female mice were treated with S-NACH or tinzaparin, and 30 min later luciferase-transfected pancreatic cancer cells (Mpanc96) were implanted into the spleen; treatment was continued daily until termination. Next we studied the effect of S-NACH versus tinzaparin on metastasis after surgical excision of the primary tumor after 3 weeks of daily treatment with S-NACH or tinzaparin. S-NACH reduced surgically induced metastasis (p<0.01) and tumor recurrence (p<0.05) relative to control. Histopathological studies demonstrated significant increase in tumor necrosis mediated by S-NACH and to lesser extent by tinzaparin as compared to control group. Furthermore, either S-NACH or tinzaparin upregulated the expression of the junctional adhesion molecule E-cadherin in pancreatic cancer cells where its low expression enhances cancer cell migration and invasion. In terms of bleeding time (BT), S-NACH did not affect BT as compared to tinzaparin, which doubled BT. These data suggest that S-NACH is an effective and safe anti-metastatic agent and warrants further clinical evaluation.
Collapse
Affiliation(s)
- Reem Alyahya
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Thangirala Sudha
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Michael Racz
- Department of Basic and Social Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Steven C Stain
- Department of Surgery, Albany Medical College, Albany, NY, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
31
|
Zhang B, Gao Z, Sun M, Li H, Fan H, Chen D, Zheng J. Prognostic significance of VEGF-C, semaphorin 3F, and neuropilin-2 expression in oral squamous cell carcinomas and their relationship with lymphangiogenesis. J Surg Oncol 2014; 111:382-8. [PMID: 25475162 DOI: 10.1002/jso.23842] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 10/18/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Bing Zhang
- Department of Anatomy; Basic Medical Science College; Harbin Medical University; Harbin China
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Harbin Medical University; Harbin China
| | - Zhongxiuzi Gao
- Department of Anatomy; Basic Medical Science College; Harbin Medical University; Harbin China
| | - Miao Sun
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Harbin Medical University; Harbin China
| | - Haixia Li
- Department of Anatomy; Basic Medical Science College; Harbin Medical University; Harbin China
| | - Haixia Fan
- Department of Anatomy; Basic Medical Science College; Harbin Medical University; Harbin China
| | - Dong Chen
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Harbin Medical University; Harbin China
| | - Jinhua Zheng
- Department of Anatomy; Basic Medical Science College; Harbin Medical University; Harbin China
| |
Collapse
|
32
|
Tawada M, Hayashi SI, Ikegame Y, Nakashima S, Yoshida K. Possible involvement of tumor-producing VEGF-A in the recruitment of lymphatic endothelial progenitor cells from bone marrow. Oncol Rep 2014; 32:2359-64. [PMID: 25242215 DOI: 10.3892/or.2014.3499] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/17/2014] [Indexed: 11/05/2022] Open
Abstract
Lymphatic metastasis of human malignant adenocarcinomas is a critical determinant of prognosis. Lymphangiogenesis, the growth of lymphatic vessels, is closely involved in lymphatic metastasis. However, the mechanisms of tumor lymphangiogenesis are not clearly understood. In a previous study, we showed that human gastric cancer MKN45 cells organize neighboring lymphatic vessels via recruitment of bone marrow-derived lymphatic endothelial progenitor cells in a nude mouse xenograft model. The present results also indicated that human colorectal cancer LS174T and breast cancer SK-BR-3 cells promoted lymphangiogenesis as well as the recruitment of lymphatic endothelial progenitor cells from bone marrow. Among growth factors, which are reported to be involved in lymphangiogenesis, only vascular endothelial growth factor (VEGF)-A was extensively secreted by these three types of adenocarcinoma cells in culture. The well-characterized lymphangiogenic factors VEGF-C and VEGF-D in the culture medium of these three types of adenocarcinoma cells were below the detectable levels in ELISA assay. Secretion of epidermal growth factor (EGF) and hepatocyte growth factor (HGF) was not detected. In in vitro culture assay, VEGF-A directly induced the differentiation of bone marrow mononuclear cells into LYVE-1-positive lymphatic endothelial lineage cells. These data collectively suggest the possibility that VEGF-A-rich human adenocarcinomas induce tumor lymphangiogenesis via recruitment of lymphangiogenic endothelial progenitor cells from bone marrow.
Collapse
Affiliation(s)
- Masahiro Tawada
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Shin-Ichiro Hayashi
- Department of Cell Signaling, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Yuka Ikegame
- Department of Cell Signaling, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Shigeru Nakashima
- Department of Cell Signaling, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| |
Collapse
|
33
|
Gogate PN, Ethirajan M, Kurenova EV, Magis AT, Pandey RK, Cance WG. Design, synthesis, and biological evaluation of novel FAK scaffold inhibitors targeting the FAK-VEGFR3 protein-protein interaction. Eur J Med Chem 2014; 80:154-166. [PMID: 24780592 DOI: 10.1016/j.ejmech.2014.04.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/10/2014] [Accepted: 04/12/2014] [Indexed: 02/08/2023]
Abstract
Focal adhesion kinase (FAK) and vascular endothelial growth factor receptor 3 (VEGFR3) are tyrosine kinases, which function as key modulators of survival and metastasis signals in cancer cells. Previously, we reported that small molecule chlorpyramine hydrochloride (C4) specifically targets the interaction between FAK and VEGFR3 and exhibits anti-tumor efficacy. In this study, we designed and synthesized a series of 1 (C4) analogs on the basis of structure activity relationship and molecular modeling. The resulting new compounds were evaluated for their binding to the FAT domain of FAK and anti-cancer activity. Amongst all tested analogs, compound 29 augmented anti-proliferative activity in multiple cancer cell lines with stronger binding to the FAT domain of FAK and disrupted the FAK-VEGFR3 interaction. In conclusion, we hope that this work will contribute to further studies of more potent and selective FAK-VEGFR3 protein-protein interaction inhibitors.
Collapse
Affiliation(s)
- Priyanka N Gogate
- Department of Cell Stress Biology/PDT Center, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Manivannan Ethirajan
- Department of Cell Stress Biology/PDT Center, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Elena V Kurenova
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,CureFAKtor Pharmaceuticals, Orchard Park, NY 14127, USA
| | - Andrew T Magis
- Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ravindra K Pandey
- Department of Cell Stress Biology/PDT Center, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - William G Cance
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,CureFAKtor Pharmaceuticals, Orchard Park, NY 14127, USA
| |
Collapse
|
34
|
Yu J, Zhang X, Kuzontkoski PM, Jiang S, Zhu W, Li DY, Groopman JE. Slit2N and Robo4 regulate lymphangiogenesis through the VEGF-C/VEGFR-3 pathway. Cell Commun Signal 2014; 12:25. [PMID: 24708522 PMCID: PMC4122147 DOI: 10.1186/1478-811x-12-25] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 02/21/2014] [Indexed: 12/28/2022] Open
Abstract
Background Signaling through vascular endothelial growth factor C (VEGF–C) and
VEGF receptor 3 (VEGFR-3) plays a central role in lymphangiogenesis and the
metastasis of several cancers via the lymphatics. Recently, the Slit2/Robo4
pathway has been recognized as a modulator of vascular permeability and
integrity. Signaling via the Robo receptor inhibits VEGF-mediated effects;
however, its effects on lymphatic endothelial cell function have not been
well characterized. Results We found that pretreatment with Slit2N, an active fragment of Slit2,
inhibited VEGF-C-mediated lung-derived lymphatic endothelial cell (L-LEC)
proliferation, migration, and in vitro tube formation. Slit2N
induced the internalization of VEGFR-3, which blocked its activation, and
inhibited the activation of the PI3K/Akt pathway by VEGF-C in L-LECs.
Moreover, we found that inhibition of VEGF-C-induced effects by Slit2N was
Robo4-dependent. Conclusion These results indicate that Slit2N/Robo4 modulates several key cellular
functions, which contribute to lymphangiogenesis, and identify this
ligand-receptor pair as a potential therapeutic target to inhibit lymphatic
metastasis of VEGF-C-overexpressing cancers and manage lymphatic
dysfunctions characterized by VEGF-C/VEGFR-3 activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jerome E Groopman
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02115, USA.
| |
Collapse
|
35
|
JAM-C promotes lymphangiogenesis and nodal metastasis in non-small cell lung cancer. Tumour Biol 2014; 35:5675-87. [PMID: 24584816 DOI: 10.1007/s13277-014-1751-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 02/13/2014] [Indexed: 01/09/2023] Open
Abstract
This study aims to investigate lymphatic metastasis-related genes in non-small cell lung carcinomas (NSCLC). NSCLC tissue was analyzed for expression of junctional adhesion molecule-C (JAM-C) protein. Our data revealed novel associations between JAM-C overexpression in primary tumors and lymphatic microvessel density (LMVD), lymph node metastasis, and poorer overall survival and recurrence-free survival. We used the highly metastatic human lung adenocarcinoma cell line Anip973 and its parental line AGZY83-a, which has a low metastatic capacity, in vivo and vitro. We found that JAM-C played an important role in different metastasis capacity of lymph node. JAM-C affected tumor growth, LNM, JAM-C, VEGF-C, vasculature, and ERK1/2 phosphorylation (p-ERK1/2). β1 integrin was involved in lymph node metastasis. Moreover, JAM-C knockdown in highly metastatic Anip973 decreased cell migration in scratch-wound assays. The JAM-C knockdown in Anip973 cells and JAM-C cDNA in AGZY83-a cells regulated the vascular endothelial growth factor C (VEGF-C) expression. Immunofluorescence showed that blocked VEGF-C expression in JAM-C shRNA Anip973 cells were restored after JAM-C treatment. JAM-C-induced VEGF-C in JAM-C cDNA AGZY83-a cells was also effectively inhibited by treatment with an antibody specifically against JAM-C. Use of media from Anip973 cells, AGZY83-a, and A549cells lung cancer cells that overexpressed or downregulated JAM-C was demonstrated to affect activity of VEGF-C-induced β1 integrin subunit or ERK activity in human dermal lymphatic endothelial cells (HDLEC) treated with VEGF-C or inhibitory antibody to JAM-C. Overall, these results indicate that JAM-C could mediate metastasis as it contributes to VEGF-C expression in cancer cells. JAM-C affects β1and ERK activation in HDLEC, thus promoting lymphangiogenesis and nodal metastasis. Our findings indicate that JAM-C may be a therapeutic target for preventing and treating lymphatic metastases.
Collapse
|
36
|
Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB, Achen MG. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer 2014; 14:159-72. [PMID: 24561443 DOI: 10.1038/nrc3677] [Citation(s) in RCA: 597] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The generation of new lymphatic vessels through lymphangiogenesis and the remodelling of existing lymphatics are thought to be important steps in cancer metastasis. The past decade has been exciting in terms of research into the molecular and cellular biology of lymphatic vessels in cancer, and it has been shown that the molecular control of tumour lymphangiogenesis has similarities to that of tumour angiogenesis. Nevertheless, there are significant mechanistic differences between these biological processes. We are now developing a greater understanding of the specific roles of distinct lymphatic vessel subtypes in cancer, and this provides opportunities to improve diagnostic and therapeutic approaches that aim to restrict the progression of cancer.
Collapse
Affiliation(s)
- Steven A Stacker
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [3] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Steven P Williams
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Tara Karnezis
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| | - Ramin Shayan
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia. [3] Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia. [4] O'Brien Institute, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Stephen B Fox
- 1] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [2] Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Marc G Achen
- 1] Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia. [3] Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
37
|
Qiu H, Cao L, Wang D, Xu H, Liang Z. High levels of circulating CD34+/VEGFR3+ lymphatic/vascular endothelial progenitor cells is correlated with lymph node metastasis in patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2013; 39:1268-75. [PMID: 23803010 DOI: 10.1111/jog.12047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 12/12/2012] [Indexed: 11/28/2022]
Abstract
AIM Lymph node metastasis is one of the predictive factors associated with poor prognosis of epithelial ovarian cancer. To clarify the role of CD34 and vascular endothelial growth factor receptor-3-positive (CD34+/VEGFR3+) lymphatic/vascular endothelial progenitor cells (LVEPC) in patients with lymph node metastasis and epithelial ovarian cancer progression, the levels of circulating CD34+/VEGFR3+ LVEPC in epithelial ovarian cancer patients were detected. We also tested the plasma protein levels of VEGF and stromal cell-derived factor to find out their possible relationships with lymph node metastasis in our epithelial ovarian cancer cohort. MATERIAL AND METHODS Peripheral blood samples were collected from 54 patients diagnosed as epithelial ovarian cancer, and 31 normal samples as control. The circulating levels of LVEPC were carried out by flow cytometry, and blood protein levels of biomarkers were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The level of circulating LVEPC was significantly higher in patients with ovarian cancer compared with that of healthy controls. There was also a statistically significant correlation between LVEPC levels and surgical staging of epithelial ovarian cancer (P < 0.01). CONCLUSION The circulating levels of bone marrow-derived LVEPC are significantly increased in epithelial ovarian cancer patients and these levels correlate with lymph node metastasis too.
Collapse
Affiliation(s)
- Huiling Qiu
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
38
|
Le C, Karnezis T, Achen MG, Stacker S, Sloan E. Lymphovascular and neural regulation of metastasis: shared tumour signalling pathways and novel therapeutic approaches. Best Pract Res Clin Anaesthesiol 2013; 27:409-25. [PMID: 24267548 PMCID: PMC4007214 DOI: 10.1016/j.bpa.2013.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
The progression of cancer is supported by a wide variety of non-neoplastic cell types which make up the tumour stroma, including immune cells, endothelial cells, cancer-associated fibroblasts and nerve fibres. These host cells contribute molecular signals that enhance primary tumour growth and provide physical avenues for metastatic dissemination. This article provides an overview of the role of blood vessels, lymphatic vessels and nerve fibres in the tumour microenvironment and highlights the interconnected molecular signalling pathways that control their development and activation in cancer. Further, this article highlights the known pharmacological agents which target these pathways and discusses the potential therapeutic uses of drugs that target angiogenesis, lymphangiogenesis and stress-response pathways in the different stages of cancer care.
Collapse
Affiliation(s)
- C.P. Le
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - T. Karnezis
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
| | - M. G. Achen
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - S.A. Stacker
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3010, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - E.K. Sloan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Cancer Anaesthesia and Pain Medicine, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
- Cousins Center for PNI, Semel Institute for Neuroscience and Human Behavior, UCLA AIDS Institute and Jonsson Comprehensive Cancer Center, University of California Los Angeles, USA
| |
Collapse
|
39
|
Hsu HW, Wall NR, Hsueh CT, Kim S, Ferris RL, Chen CS, Mirshahidi S. Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol 2013; 50:19-26. [PMID: 24269532 DOI: 10.1016/j.oraloncology.2013.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/24/2013] [Accepted: 10/02/2013] [Indexed: 02/02/2023]
Abstract
Tumor angiogenesis is a hallmark of advanced cancers and promotes invasion and metastasis. Over 90% of head and neck squamous cell carcinomas (HNSCC) express angiogenic factors such as vascular endothelial growth factor (VEGF). Several preclinical studies support the prognostic implications of angiogenic markers for HNSCC and currently this is an attractive treatment target in solid tumors. Since radiotherapy is one of the most commonly used treatments for HNSCC, it is imperative to identify the interactions between antiangiogenic therapy and radiotherapy, and to develop combination therapy to improve clinical outcome. The mechanisms between antiangiogenic agents and ionizing radiation are complicated and involve many interactions between the vasculature, tumor stroma and tumor cells. The proliferation and metastasis of tumor cells rely on angiogenesis/blood vessel formation. Rapid growing tumors will cause hypoxia, which up-regulates tumor cell survival factors, such as hypoxia-inducing factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), giving rise to more tumor proliferation, angiogenesis and increased radioresistance. Thus, agents that target tumor vasculature and new tumor vessel formation can modulate the tumor microenvironment to improve tumor blood flow and oxygenation, leading to enhanced radiosensitivity. In this review, we discuss the mechanisms of how antiangiogenic therapies improve tumor response to radiation and data that support this combination strategy as a promising method for the treatment of HNSCC in the future.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Department of Pharmacology, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA
| | - Nathan R Wall
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA, USA
| | - Chung-Tsen Hsueh
- Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Seungwon Kim
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien-Shing Chen
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA; Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Saied Mirshahidi
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
40
|
Rutkowski JM, Ihm JE, Lee ST, Kilarski WW, Greenwood VI, Pasquier MC, Quazzola A, Trono D, Hubbell JA, Swartz MA. VEGFR-3 neutralization inhibits ovarian lymphangiogenesis, follicle maturation, and murine pregnancy. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1596-1607. [PMID: 24036251 DOI: 10.1016/j.ajpath.2013.07.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/15/2013] [Accepted: 07/31/2013] [Indexed: 11/18/2022]
Abstract
Lymphatic vessels surround follicles within the ovary, but their roles in folliculogenesis and pregnancy, as well as the necessity of lymphangiogenesis in follicle maturation and health, are undefined. We used systemic delivery of mF4-31C1, a specific antagonist vascular endothelial growth factor receptor 3 (VEGFR-3) antibody to block lymphangiogenesis in mice. VEGFR-3 neutralization for 2 weeks before mating blocked ovarian lymphangiogenesis at all stages of follicle maturation, most notably around corpora lutea, without significantly affecting follicular blood angiogenesis. The numbers of oocytes ovulated, fertilized, and implanted in the uterus were normal in these mice; however, pregnancies were unsuccessful because of retarded fetal growth and miscarriage. Fewer patent secondary follicles were isolated from treated ovaries, and isolated blastocysts exhibited reduced cell densities. Embryos from VEGFR-3-neutralized dams developed normally when transferred to untreated surrogates. Conversely, normal embryos transferred into mF4-31C1-treated dams led to the same fetal deficiencies observed with in situ gestation. Although no significant changes were measured in uterine blood or lymphatic vascular densities, VEGFR-3 neutralization reduced serum and ovarian estradiol concentrations during gestation. VEGFR-3-mediated lymphangiogenesis thus appears to modulate the folliculogenic microenvironment and may be necessary for maintenance of hormone levels during pregnancy; both of these are novel roles for the lymphatic vasculature.
Collapse
Affiliation(s)
- Joseph M Rutkowski
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Jong Eun Ihm
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Seung Tae Lee
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Witold W Kilarski
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Veronique I Greenwood
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Miriella C Pasquier
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Alexandra Quazzola
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Didier Trono
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Jeffrey A Hubbell
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Melody A Swartz
- Institute of Bioengineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
41
|
Vascular remodeling in cancer. Oncogene 2013; 33:3496-505. [PMID: 23912450 DOI: 10.1038/onc.2013.304] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 05/31/2013] [Accepted: 06/10/2013] [Indexed: 12/17/2022]
Abstract
The growth and dissemination of tumors rely on an altered vascular network, which supports their survival and expansion and provides accessibility to the vasculature and a route of transport for metastasizing tumor cells. The remodeling of vascular structures through generation of new vessels (for example, via tumor angiogenesis) is a well studied, even if still quite poorly understood, process in human cancer. Antiangiogenic therapies have provided insight into the contribution of angiogenesis to the biology of human tumors, yet have also revealed the ease with which resistance to antiangiogenic drugs can develop, presumably involving alterations to vascular signaling mechanisms. Furthermore, cellular and/or molecular changes to pre-existing vessels could represent subtle pre-metastatic alterations to the vasculature, which are important for cancer progression. These changes, and associated molecular markers, may forecast the behavior of individual tumors and contribute to the early detection, diagnosis and prognosis of cancer. This review, which primarily focuses on the blood vasculature, explores current knowledge of how tumor vessels can be remodeled, and the cellular and molecular events responsible for this process.
Collapse
|
42
|
Vascular Endothelial Growth Factor-d Modulates Caliber and Function of Initial Lymphatics in the Dermis. J Invest Dermatol 2013; 133:2074-84. [DOI: 10.1038/jid.2013.83] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 12/21/2022]
|
43
|
Dębiński P, Dembowski J, Kowal P, Szydełko T, Kołodziej A, Małkiewicz B, Tupikowski K, Zdrojowy R. The clinical significance of lymphangiogenesis in renal cell carcinoma. Med Sci Monit 2013; 19:606-11. [PMID: 23881345 PMCID: PMC3724570 DOI: 10.12659/msm.883981] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The formation of lymphatic vessels (lymphangiogenesis) occurs in tumor tissues and is crucial for tumor development and progression in some cancers. Lymphangiogenesis and its clinical effect on renal cell carcinoma have been less thoroughly investigated in comparison with angiogenesis. The aim of this study was to evaluate the role of lymphangiogenesis as a prognostic factor in renal cell carcinoma (RCC). Material/Methods The expression of peritumoral/intratumoral lymphatics was studied by immunohistochemical methods in paraffin-embedded nephrectomy specimens from 133 patients with clear cell carcinoma. Patients were divided into 3 groups depending on postoperative follow-up: I) patients without metastases, II) patients with metastases during follow-up, and III) patients with metastases during the operation. Peritumoral lymphatics (PTL) and intratumoral lymphatics (ITL) were immunostained with a D2-40 antibody. Results The mean number of PTL present in each group was I=14.1, II=10.6, III=12.1. The mean number of ITL present in each group was I=0.7, II=2.3, III=2.3. The 3 groups showed statistically significant differences only in the case of ITL. A mean count of ITL ≥1 is significantly associated with an increased risk of regional lymph node involvement and distant metastasis. Patients with expression ITL >0.2 and PTL ≤15.2 had a significantly shorter cancer-specific survival. Conclusions The number of ITL showed an association with more aggressive cases of RCC and progression of disease. Therefore, the level of expression ITL, together with stage and histological grading, may provide valuable predictive information about the outcome of treatment.
Collapse
Affiliation(s)
- Paweł Dębiński
- Clinic of Urology and Oncological Urology, Wrocław Medical University, Wrocław, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Signaling for lymphangiogenesis via VEGFR-3 is required for the early events of metastasis. Clin Exp Metastasis 2013; 30:819-32. [PMID: 23591595 DOI: 10.1007/s10585-013-9581-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 04/02/2013] [Indexed: 01/08/2023]
Abstract
Metastasis to regional lymph nodes is an important and early event in many tumors. Vascular endothelial growth factor-C (VEGF-C), VEGF-D and their receptor VEGFR-3, play a role in tumor spread via the lymphatics, although the timing of their involvement is not understood. In contrast, VEGFR-2, activated by VEGF-A, VEGF-C and VEGF-D, is a mediator of angiogenesis and drives primary tumor growth. We demonstrate the critical role for VEGFR-3, but not VEGFR-2, in the early events of metastasis. In a tumor model exhibiting both VEGF-D-dependent angiogenesis and lymphangiogenesis, an antibody to VEGFR-2 (DC101) was capable of inhibiting angiogenesis (79 % reduction in PECAM + blood vessels) and growth (93 % reduction in tumor volume). However, unlike an anti-VEGFR-3 Mab (mF4-31C1), DC101 was not capable of eliminating either tumor lymphangiogenesis or lymphogenous metastasis (60 % reduction of lymph node metastasis by DC101 vs 95 % by mF4-31C1). Early excision of the primary tumors demonstrated that VEGF-D-mediated tumor spread precedes angiogenesis-induced growth. Small but highly metastatic primary human breast cancers had significantly higher lymphatic vessel density (23.1 vessels/mm(2)) than size-matched (11.7) or larger non-metastatic tumors (12.4) thus supporting the importance of lymphatic vessels, as opposed to angiogenesis-mediated primary tumor growth, for nodal metastasis. These results suggest that lymphangiogenesis via VEGF-D is more critical than angiogenesis for nodal metastasis.
Collapse
|
45
|
Sun JJ, Jing W, Ni YY, Yuan XJ, Zhou HH, Fan YZ. New model of in-situ xenograft lymphangiogenesis by a human colonic adenocarcinoma cell line in nude mice. Asian Pac J Cancer Prev 2013; 13:2823-8. [PMID: 22938466 DOI: 10.7314/apjcp.2012.13.6.2823] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To explore a new model of in-situ xenograft lymphangiogenesis of human colonic adenocarcinomas in nude mice. METHOD On the basis of establishing subcutaneous xenograft lymphangiogenesis model of human colonic adenocarcinoms, in-situ xenografts were established through the in situ growth of the HT-29 human colonic adenocarcinoma cell line in nude mice. The numbers of lymphangiogenic microvessels, the expression of lymphatic endothelial cell markers lymphatic vessel endothelial hyaloronic acid receptor-1 (LYVE-1), D2-40 and the lymphatic endothelial growth factors vascular endothelial growth factor-C (VEGF-C), -D (VEGF-D) and receptor-3 (VEGFR-3) were compared by immunohistochemical staining, Western bolt and quantitative RT-PCR in xenograft in-situ models. RESULTS Some microlymphatics with thin walls, large and irregular or collapsed cavities and increased LMVD, with strong positive of LYVE-1, D2-40 in immunohistochemistry, were observed, identical with the morphological characteristics of lymphatic vessels and capillaries. Expression of LYVE-1 and D2-40 proteins and mRNAs were significantly higher in xenografts in-situ than in the negative control group (both P<0.01). Moreover, the expression of VEGF-C, VEGF-D and VEGFR-3 proteins and mRNAs were significantly higher in xenografts in-situ (both P<0.01), in conformity with the signal regulation of the VEGF-C,-D/VEGFR-3 axis of tumor lymphangiogenesis. CONCLUSIONS In-situ xenografts of a human colonic adenocarcinoma cell line demonstrate tumor lymphangiogenesis. This novel in-situ animal model should be useful for further studying mechanisms of lymph node metastasis, drug intervention and anti-metastasis therapy in colorectal cancer.
Collapse
Affiliation(s)
- Jian-Jun Sun
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
46
|
Stacker SA, Achen MG. The VEGF signaling pathway in cancer: the road ahead. CHINESE JOURNAL OF CANCER 2013; 32:297-302. [PMID: 23419196 PMCID: PMC3845619 DOI: 10.5732/cjc.012.10319] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF) family of soluble protein growth factors consists of key mediators of angiogenesis and lymphangiogenesis in the context of tumor biology. The members of the family, VEGF-A (also known as VEGF), VEGF-B, VEGF-C, VEGF-D, and placenta growth factor (PIGF), play important roles in vascular biology in both normal physiology and pathology. The generation of a humanized neutralizing antibody to VEGF-A (bevacizumab, also known as Avastin) and the demonstration of its benefit in numerous human cancers have confirmed the merit of an anti-angiogenesis approach to cancer treatment and have validated the VEGF-A signaling pathway as a therapeutic target. Other members of the VEGF family are now being targeted, and their relevance to human cancer and the development of resistance to anti-VEGF-A treatment are being evaluated in the clinic. Here, we discuss the potential of targeting VEGF family members in the diagnosis and treatment of cancer.
Collapse
|
47
|
Harris NC, Davydova N, Roufail S, Paquet-Fifield S, Paavonen K, Karnezis T, Zhang YF, Sato T, Rothacker J, Nice EC, Stacker SA, Achen MG. The propeptides of VEGF-D determine heparin binding, receptor heterodimerization, and effects on tumor biology. J Biol Chem 2013; 288:8176-8186. [PMID: 23404505 DOI: 10.1074/jbc.m112.439299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
VEGF-D is an angiogenic and lymphangiogenic glycoprotein that can be proteolytically processed generating various forms differing in subunit composition due to the presence or absence of N- and C-terminal propeptides. These propeptides flank the central VEGF homology domain, that contains the binding sites for VEGF receptors (VEGFRs), but their biological functions were unclear. Characterization of propeptide function will be important to clarify which forms of VEGF-D are biologically active and therefore clinically relevant. Here we use VEGF-D mutants deficient in either propeptide, and in the capacity to process the remaining propeptide, to monitor the functions of these domains. We report for the first time that VEGF-D binds heparin, and that the C-terminal propeptide significantly enhances this interaction (removal of this propeptide from full-length VEGF-D completely prevents heparin binding). We also show that removal of either the N- or C-terminal propeptide is required for VEGF-D to drive formation of VEGFR-2/VEGFR-3 heterodimers which have recently been shown to positively regulate angiogenic sprouting. The mature form of VEGF-D, lacking both propeptides, can also promote formation of these receptor heterodimers. In a mouse tumor model, removal of only the C-terminal propeptide from full-length VEGF-D was sufficient to enhance angiogenesis and tumor growth. In contrast, removal of both propeptides is required for high rates of lymph node metastasis. The findings reported here show that the propeptides profoundly influence molecular interactions of VEGF-D with VEGF receptors, co-receptors, and heparin, and its effects on tumor biology.
Collapse
Affiliation(s)
- Nicole C Harris
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Natalia Davydova
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Sally Roufail
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Sophie Paquet-Fifield
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Karri Paavonen
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Tara Karnezis
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - You-Fang Zhang
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Teruhiko Sato
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Julie Rothacker
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Edouard C Nice
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Steven A Stacker
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3050, Australia
| | - Marc G Achen
- Tumour Angiogenesis Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia; Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3050, Australia.
| |
Collapse
|
48
|
Zhao G, Rodriguez BL. Molecular targeting of liposomal nanoparticles to tumor microenvironment. Int J Nanomedicine 2012; 8:61-71. [PMID: 23293520 PMCID: PMC3534304 DOI: 10.2147/ijn.s37859] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Liposomes are biodegradable and can be used to deliver drugs at a much higher concentration in tumor tissues than in normal tissues. Both passive and active drug delivery by liposomal nanoparticles can significantly reduce the toxic side effects of anticancer drugs and enhance the therapeutic efficacy of the drugs delivered. Active liposomal targeting to tumors is achieved by recognizing specific tumor receptors through tumor-specific ligands or antibodies coupled onto the surface of the liposomes, or by stimulus-sensitive drug carriers such as acid-triggered release or enzyme-triggered drug release. Tumors are often composed of tumor cells and nontumor cells, which include endothelial cells, pericytes, fibroblasts, stromal, mesenchymal cells, innate, and adaptive immune cells. These nontumor cells thus form the tumor microenvironment, which could be targeted and modified so that it is unfavorable for tumor cells to grow. In this review, we briefly summarized articles that had taken advantage of liposomal nanoparticles as a carrier to deliver anticancer drugs to the tumor microenvironment, and how they overcame obstacles such as nonspecific uptake, interaction with components in blood, and toxicity. Special attention is devoted to the liposomal targeting of anticancer drugs to the endothelium of tumor neovasculature, tumor associated macrophages, fibroblasts, and pericytes within the tumor microenvironment.
Collapse
Affiliation(s)
- Gang Zhao
- Institute of Materia Medica, Shandong Academy of Medical Science, Shandong, China
| | | |
Collapse
|
49
|
Shibata MA, Ambati J, Shibata E, Yoshidome K, Harada-Shiba M. Mammary cancer gene therapy targeting lymphangiogenesis: VEGF-C siRNA and soluble VEGF receptor-2, a splicing variant. Med Mol Morphol 2012; 45:179-84. [PMID: 23224595 DOI: 10.1007/s00795-012-0576-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/14/2012] [Indexed: 11/26/2022]
Abstract
Metastasis contributes significantly to cancer mortality, and the most common pathway of initial dissemination is via the afferent ducts of the lymphatics. Overexpression of vascular endothelial growth factor (VEGF)-C has been associated with lymphangiogenesis and lymph node metastasis in a multitude of human neoplasms, including breast cancers. We recently reported that both VEGF-C siRNA and endogenous soluble vascular endothelial growth factor receptor-2 (esVEGFR-2, a new splicing variant) inhibit VEGF-C function and metastasis in a mouse model of metastatic mammary cancer. Here we briefly review our previous experimental work, specifically targeting tumor lymphangiogenesis, in which metastatic mouse mammary cancers received direct intratumoral injections of either expression vectors VEGF-C siRNA or esVEGFR-2, or the empty plasmid vector, once a week for 6 or 8 weeks, followed by in vivo gene electrotransfer of the injected tumors. Throughout our study, both tumor lymphangiogenesis and the multiplicity of lymph node metastasis were significantly inhibited, with an overall reduction in tumor growth, by both VEGF-C siRNA and esVEGFR-2; further, a significant reduction in the number of dilated lymphatic vessels containing intraluminal cancer cells was observed with both treatments. Thus, therapeutic strategies targeting lymphangiogenesis may have great clinical significance for the treatment of metastatic human breast cancer.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Laboratory of Anatomy and Histopathology, Faculty of Health Science, Osaka Health Science University, 1-9-27 Temma, Osaka 530-0043, Japan.
| | | | | | | | | |
Collapse
|
50
|
Kesler CT, Liao S, Munn LL, Padera TP. Lymphatic vessels in health and disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2012. [PMID: 23209022 DOI: 10.1002/wsbm.1201] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The lymphatic vasculature plays vital roles in tissue fluid balance, immune defense, metabolism, and cancer metastasis. In adults, lymphatic vessel formation and remodeling occur primarily during inflammation, development of the corpus luteum, wound healing, and tumor growth. Unlike the blood circulation, where unidirectional flow is sustained by the pumping actions of the heart, pumping actions intrinsic to the lymphatic vessels themselves are important drivers of lymphatic flow. This review summarizes critical components that control lymphatic physiology.
Collapse
Affiliation(s)
- Cristina T Kesler
- E. L. Steele Laboratory, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | | | | | | |
Collapse
|