101
|
Novel anti-angiogenic effects of aromatic-turmerone, essential oil isolated from spice turmeric. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
102
|
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
|
103
|
Morton JJ, Bird G, Keysar SB, Astling DP, Lyons TR, Anderson RT, Glogowska MJ, Estes P, Eagles JR, Le PN, Gan G, McGettigan B, Fernandez P, Padilla-Just N, Varella-Garcia M, Song JI, Bowles DW, Schedin P, Tan AC, Roop DR, Wang XJ, Refaeli Y, Jimeno A. XactMice: humanizing mouse bone marrow enables microenvironment reconstitution in a patient-derived xenograft model of head and neck cancer. Oncogene 2015; 35:290-300. [PMID: 25893296 PMCID: PMC4613815 DOI: 10.1038/onc.2015.94] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 12/14/2022]
Abstract
The limitations of cancer cell lines have led to the development of direct patient derived xenograft (PDX) models. However, the interplay between the implanted human cancer cells and recruited mouse stromal and immune cells alters the tumor microenvironment and limits the value of these models. To overcome these constraints, we have developed a technique to expand human hematopoietic stem and progenitor cells (HSPCs) and use them to reconstitute the radiation-depleted bone marrow of a NOD/SCID/IL2rg−/− (NSG) mouse on which a patient’s tumor is then transplanted (XactMice). The human HSPCs produce immune cells that home into the tumor and help replicate its natural microenvironment. Despite previous passage on nude mice, the expression of epithelial, stromal, and immune genes in XactMice tumors aligns more closely to that of the patient tumor than to those grown in non-humanized mice – an effect partially facilitated by human cytokines expressed by both the HSPC progeny and the tumor cells. The human immune and stromal cells produced in the XactMice can help recapitulate the microenvironment of an implanted xenograft, reverse the initial genetic drift seen after passage on non-humanized mice, and provide a more accurate tumor model to guide patient treatment.
Collapse
Affiliation(s)
- J J Morton
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - G Bird
- Department of Dermatology, University of Colorado School of Medicine
| | - S B Keysar
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - D P Astling
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Biostatistics and Informatics, University of Colorado School of Medicine
| | - T R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - R T Anderson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - M J Glogowska
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Estes
- Department of Dermatology, University of Colorado School of Medicine
| | - J R Eagles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P N Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - G Gan
- Department of Radiation Oncology, University of Colorado School of Medicine
| | - B McGettigan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Fernandez
- Department of Pathology, University of Colorado School of Medicine
| | - N Padilla-Just
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - M Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - J I Song
- Department of Otolaryngology, University of Colorado School of Medicine
| | - D W Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Schedin
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - A-C Tan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Biostatistics and Informatics, University of Colorado School of Medicine
| | - D R Roop
- Department of Dermatology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - X-J Wang
- Department of Pathology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Y Refaeli
- Department of Dermatology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - A Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Otolaryngology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
104
|
Maeng YS, Aguilar B, Choi SI, Kim EK. Inhibition of TGFBIp expression reduces lymphangiogenesis and tumor metastasis. Oncogene 2015; 35:196-205. [PMID: 25772247 DOI: 10.1038/onc.2015.73] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/17/2014] [Accepted: 02/05/2015] [Indexed: 01/18/2023]
Abstract
Transforming growth factor-β-induced protein (TGFBIp) is an extracellular matrix protein that has a role in a wide range of pathological conditions. However, the role of TGFBIp signaling in lymphangiogenesis is poorly understood. The purpose of this study was therefore to analyze the effects of TGFBIp on lymphangiogenesis and determine whether TGFBIp-related lymphangiogenesis is important for the metastasis of tumor cells. TGFBIp increased adhesion, migration, and morphologic differentiation of human lymphatic endothelial cells (LECs), consistent with an increase in lymphatic vessel sprouting in a three-dimensional lymphatic ring assay. TGFBIp also induced phosphorylation of intracellular signaling molecules SRC, FAK, AKT, JNK and ERK. TGFBIp-induced lymphatic vessel sprouting was inhibited by addition of anti-integrin β3 antibody and pharmacologic inhibitors of FAK, AKT, JNK or ERK. TGFBIp increased both CCL21 expression in LECs, a chemokine that actively recruits tumor cells expressing the cognate chemokine receptors to lymphatic vessels and LEC permeability by inducing the dissociation of VE-cadherin junctions between LECs via the activation of SRC signaling. In vivo, inhibition of TGFBIp expression in SW620 cancer cells dramatically reduced tumor lymphangiogenesis and metastasis. Collectively, our findings demonstrate that TGFBIp is a lymphangiogenic factor contributing to tumor dissemination and represents a potential target to inhibit metastasis.
Collapse
Affiliation(s)
- Y-S Maeng
- Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - B Aguilar
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA
| | - S-I Choi
- Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - E K Kim
- Corneal Dystrophy Research Institute, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
105
|
Cho S, Choi YS, Yun BH, Chon SJ, Jung YS, Kim HY, Park JH, Seo SK, Kim SH, Lee BS. Effects of levonorgestrel-releasing intrauterine system on lymphangiogenesis of adenomyosis. Am J Clin Pathol 2015; 143:352-61. [PMID: 25696793 DOI: 10.1309/ajcpp8f4sfyfvxrn] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES Lymphangiogenesis may be involved in the pathogenesis of adenomyosis. We investigated the lymphatic vessels of patients with adenomyosis, including those treated with levonorgestrel-releasing intrauterine system (LNG-IUS). METHODS Full-thickness uterine samples were obtained from patients who received hysterectomies. Twenty-one patients with adenomyosis and 17 patients with adenomyosis who were treated with LNG-IUS were included. Eighteen patients with cervical intraepithelial neoplasia served as controls. Immunohistochemical staining was performed with antibodies against podoplanin and lymphatic vessel endothelial hyaluronan receptor 1. The lymphovascular density (LVD) was analyzed in each sample by the "hot spot" method. RESULTS The LVDs were significantly higher in the endometrial and myometrial tissues of patients with adenomyosis compared with those of patients treated with the LNG-IUS or controls. No significant differences were noted between the LNG-IUS-treated group and controls. Evaluation of the LVDs according to the menstrual cycle showed that the differences in the endometrial tissues of the adenomyosis group and those of the LNG-IUS-treated group or the controls were more prominent during the secretory phase. CONCLUSIONS Treatment with the LNG-IUS resulted in reduced lymphangiogenesis and LVD in the endometrial and myometrial tissues of patients with adenomyosis. Reduced lymphangiogenesis may be one mechanism by which the LNG-IUS reduces adenomyosis-related symptoms.
Collapse
Affiliation(s)
- SiHyun Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sik Choi
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Hyon Yun
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Joo Chon
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yeon Soo Jung
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Yeon Kim
- Department of Obstetrics and Gynecology, YongIn Severance Hospital, Yonsei University College of Medicine, YongIn City, Kyunggi-do, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Hyun Park
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Kyo Seo
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Byung Seok Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
106
|
The role of CCL21/CCR7 chemokine axis in breast cancer-induced lymphangiogenesis. Mol Cancer 2015; 14:35. [PMID: 25744065 PMCID: PMC4339430 DOI: 10.1186/s12943-015-0306-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 01/27/2015] [Indexed: 11/29/2022] Open
Abstract
Background Tumor-induced lymphangiogenesis facilitates breast cancer progression by generating new lymphatic vessels that serve as conduits for tumor dissemination to lymph nodes and beyond. Given the recent evidence suggesting the implication of C-C chemokine ligand 21/chemokine receptor 7 (CCL21/CCR7) in lymph node metastasis, the aim of our study was to define the role of this chemokine pair in breast cancer-associated lymphangiogenesis. Methods The expression analysis of CCL21/CCR7 pair and lymphatic endothelial cell (LEC) markers in breast cancer specimens was performed by means of quantitative real-time PCR. By utilizing CCR7 and CCL21 gene manipulated breast cancer cell implants into orthotopic sites of nude mice, lymphatic vessel formation was assessed through quantitative real-time PCR, immunohistochemistry and immunofluorescence assays. Finally, the lymphangiogenic potential of CCL21/CCR7 was assessed in vitro with primary LECs through separate functional assays, each attempting to mimic different stages of the lymphangiogenic process. Results We found that CCR7 mRNA expression in human breast cancer tissues positively correlates with the expression of lymphatic endothelial markers LYVE-1, podoplanin, Prox-1, and vascular endothelial growth factor-C (VEGF-C). We demonstrated that the expression of CCL21/CCR7 by breast cancer cells has the ability to promote tumor-induced lymph-vascular recruitment in vivo. In vitro, CCL21/CCR7 chemokine axis regulates the expression and secretion of lymphangiogenic factor VEGF-C and thereby promotes proliferation, migration, as well as tube formation of the primary human LECs. Finally, we showed that protein kinase B (AKT) signaling pathway is the intracellular mechanism of CCR7-mediated VEGF-C secretion by human breast cancer cells. Conclusions These results reveal that CCR7 and VEGF-C display a significant crosstalk and suggest a novel role of the CCL21/CCR7 chemokine axis in the promotion of breast cancer-induced lymphangiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0306-4) contains supplementary material, which is available to authorized users.
Collapse
|
107
|
Patsouras D, Papaxoinis K, Kostakis A, Safioleas MC, Lazaris AC, Nicolopoulou-Stamati P. Fibroblast activation protein and its prognostic significance in correlation with vascular endothelial growth factor in pancreatic adenocarcinoma. Mol Med Rep 2015; 11:4585-90. [PMID: 25625587 DOI: 10.3892/mmr.2015.3259] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 11/20/2014] [Indexed: 11/05/2022] Open
Abstract
Fibroblast activation protein (FAP), a selective protein for tumor stromal fibroblasts, is expressed in >90% of human epithelial carcinomas. A characteristic feature of pancreatic cancer is an extensive fibrotic or desmoplastic reaction surrounding the primary tumor. The present study aimed to evaluate the expression levels of FAP and vascular endothelial growth factor (VEGF) and determine their correlation in pancreatic adenocarcinoma. Confocal laser scanning microscopy and conventional immunohistochemical analysis were used to quantify FAP and VEGF expression levels in formalin‑fixed and paraffin‑embedded tissue biopsies from 46 patients (male, 26; female, 20; mean age, 66 years; age range, 53‑80 years) with pancreatic adenocarcinoma stage IIA or IIB. The expression levels of FAP in the neoplastic and adjacent normal tissue were significantly higher in stage IIB patients, compared with stage IIA patients. FAP expression was correlated with positive lymph nodes, resulting in poor prognosis for stage IIB patients. The partial correlation coefficient between FAP and VEGF expression levels was 0.39 (P=0.007), and the two factors had an effect on patient survival. Multivariate analysis demonstrated the prognostic superiority of FAP over VEGF, which is considered to be the most consistently reproducible molecular marker with prognostic value in resected pancreatic adenocarcinoma. Due to the limited beneficial effect of current systemic therapies for pancreatic adenocarcinoma, targeting FAP may be a potential therapeutic strategy and requires further investigation.
Collapse
Affiliation(s)
- Dimitrios Patsouras
- Department of GI Surgery, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Kostis Papaxoinis
- Gastroenterology Unit, 1st Department of Internal Medicine‑Propaedeutic, 'Laiko' General Hospital, Athens University Medical School, Athens GR‑11527, Greece
| | - Alkiviadis Kostakis
- Center of Experimental Surgery, Biomedical Research Foundation, Academy of Athens, Athens GR‑11527, Greece
| | - Michael C Safioleas
- Fourth Propedeutic Department of Surgery, Athens University Medical School, Attikon Hospital of Athens, Athens GR-12462, Greece
| | - Andreas C Lazaris
- 1st Department of Pathology, Athens University Medical School, Athens GR‑11527, Greece
| | | |
Collapse
|
108
|
Wang Z, Dabrosin C, Yin X, Fuster MM, Arreola A, Rathmell WK, Generali D, Nagaraju GP, El-Rayes B, Ribatti D, Chen YC, Honoki K, Fujii H, Georgakilas AG, Nowsheen S, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich B, Yang X, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Halicka D, Mohammed SI, Azmi AS, Bilsland A, Keith WN, Jensen LD. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 2015; 35 Suppl:S224-S243. [PMID: 25600295 PMCID: PMC4737670 DOI: 10.1016/j.semcancer.2015.01.001] [Citation(s) in RCA: 329] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 12/25/2014] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
Deregulation of angiogenesis – the growth of new blood vessels from an existing vasculature – is a main driving force in many severe human diseases including cancer. As such, tumor angiogenesis is important for delivering oxygen and nutrients to growing tumors, and therefore considered an essential pathologic feature of cancer, while also playing a key role in enabling other aspects of tumor pathology such as metabolic deregulation and tumor dissemination/metastasis. Recently, inhibition of tumor angiogenesis has become a clinical anti-cancer strategy in line with chemotherapy, radiotherapy and surgery, which underscore the critical importance of the angiogenic switch during early tumor development. Unfortunately the clinically approved anti-angiogenic drugs in use today are only effective in a subset of the patients, and many who initially respond develop resistance over time. Also, some of the anti-angiogenic drugs are toxic and it would be of great importance to identify alternative compounds, which could overcome these drawbacks and limitations of the currently available therapy. Finding “the most important target” may, however, prove a very challenging approach as the tumor environment is highly diverse, consisting of many different cell types, all of which may contribute to tumor angiogenesis. Furthermore, the tumor cells themselves are genetically unstable, leading to a progressive increase in the number of different angiogenic factors produced as the cancer progresses to advanced stages. As an alternative approach to targeted therapy, options to broadly interfere with angiogenic signals by a mixture of non-toxic natural compound with pleiotropic actions were viewed by this team as an opportunity to develop a complementary anti-angiogenesis treatment option. As a part of the “Halifax Project” within the “Getting to know cancer” framework, we have here, based on a thorough review of the literature, identified 10 important aspects of tumor angiogenesis and the pathological tumor vasculature which would be well suited as targets for anti-angiogenic therapy: (1) endothelial cell migration/tip cell formation, (2) structural abnormalities of tumor vessels, (3) hypoxia, (4) lymphangiogenesis, (5) elevated interstitial fluid pressure, (6) poor perfusion, (7) disrupted circadian rhythms, (8) tumor promoting inflammation, (9) tumor promoting fibroblasts and (10) tumor cell metabolism/acidosis. Following this analysis, we scrutinized the available literature on broadly acting anti-angiogenic natural products, with a focus on finding qualitative information on phytochemicals which could inhibit these targets and came up with 10 prototypical phytochemical compounds: (1) oleanolic acid, (2) tripterine, (3) silibinin, (4) curcumin, (5) epigallocatechin-gallate, (6) kaempferol, (7) melatonin, (8) enterolactone, (9) withaferin A and (10) resveratrol. We suggest that these plant-derived compounds could be combined to constitute a broader acting and more effective inhibitory cocktail at doses that would not be likely to cause excessive toxicity. All the targets and phytochemical approaches were further cross-validated against their effects on other essential tumorigenic pathways (based on the “hallmarks” of cancer) in order to discover possible synergies or potentially harmful interactions, and were found to generally also have positive involvement in/effects on these other aspects of tumor biology. The aim is that this discussion could lead to the selection of combinations of such anti-angiogenic compounds which could be used in potent anti-tumor cocktails, for enhanced therapeutic efficacy, reduced toxicity and circumvention of single-agent anti-angiogenic resistance, as well as for possible use in primary or secondary cancer prevention strategies.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Daniele Generali
- Molecular Therapy and Pharmacogenomics Unit, AO Isituti Ospitalieri di Cremona, Cremona, Italy
| | - Ganji P Nagaraju
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, USA
| | - Kanya Honoki
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirate University, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirate University, United Arab Emirates
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guilford, Surrey, UK
| | | | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Asfar S Azmi
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lasse D Jensen
- Department of Medical, and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
109
|
Biological ablation of sentinel lymph node metastasis in submucosally invaded early gastrointestinal cancer. Mol Ther 2014; 23:501-9. [PMID: 25523761 DOI: 10.1038/mt.2014.244] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/14/2014] [Indexed: 12/16/2022] Open
Abstract
Currently, early gastrointestinal cancers are treated endoscopically, as long as there are no lymph node metastases. However, once a gastrointestinal cancer invades the submucosal layer, the lymph node metastatic rate rises to higher than 10%. Therefore, surgery is still the gold standard to remove regional lymph nodes containing possible metastases. Here, to avoid prophylactic surgery, we propose a less-invasive biological ablation of lymph node metastasis in submucosally invaded gastrointestinal cancer patients. We have established an orthotopic early rectal cancer xenograft model with spontaneous lymph node metastasis by implantation of green fluorescent protein (GFP)-labeled human colon cancer cells into the submucosal layer of the murine rectum. A solution containing telomerase-specific oncolytic adenovirus was injected into the peritumoral submucosal space, followed by excision of the primary rectal tumors mimicking the endoscopic submucosal dissection (ESD) technique. Seven days after treatment, GFP signals had completely disappeared indicating that sentinel lymph node metastasis was selectively eradicated. Moreover, biologically treated mice were confirmed to be relapse-free even 4 weeks after treatment. These results indicate that virus-mediated biological ablation selectively targets lymph node metastasis and provides a potential alternative to surgery for submucosal invasive gastrointestinal cancer patients.
Collapse
|
110
|
Santanelli di Pompeo F, Laporta R, Longo B, Sorotos M, Papa A. Lymphatic drainage study after latissimus dorsi flap breast reconstruction. EUROPEAN JOURNAL OF PLASTIC SURGERY 2014. [DOI: 10.1007/s00238-014-0997-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
111
|
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: 14] [Impact Index Per Article: 1.3] [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
|
112
|
Shi K, Queiroz KCS, Roelofs JJTH, van Noesel CJM, Richel DJ, Spek CA. Protease-activated receptor 2 suppresses lymphangiogenesis and subsequent lymph node metastasis in a murine pancreatic cancer model. J Pathol 2014; 234:398-409. [DOI: 10.1002/path.4411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/14/2014] [Accepted: 07/18/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Kun Shi
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| | - Karla CS Queiroz
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| | - Joris JTH Roelofs
- Department of Pathology; Academic Medical Centre; Amsterdam The Netherlands
| | | | - Dirk J Richel
- Department of Medical Oncology; Academic Medical Centre; Amsterdam The Netherlands
| | - C Arnold Spek
- Centre for Experimental and Molecular Medicine; Academic Medical Centre; Amsterdam The Netherlands
| |
Collapse
|
113
|
Chen GQ, Tian H, Yue WM, Li L, Li SH, Qi L, Gao C, Si LB, Lu M, Feng F. SIRT1 expression is associated with lymphangiogenesis, lymphovascular invasion and prognosis in pN0 esophageal squamous cell carcinoma. Cell Biosci 2014; 4:48. [PMID: 25922660 PMCID: PMC4412293 DOI: 10.1186/2045-3701-4-48] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/09/2014] [Indexed: 12/15/2022] Open
Abstract
Background Sirtuin1 (SIRT1) is an NAD+-dependent type III histone deacetylase (HDAC). This research investigated the prevalence of SIRT1 protein expression and its prognostic influence with the aim of validating its potential role in lymphangiogenesis and lymphovascular invasion (LVI) in pN0 esophageal squamous cell carcinoma (ESCC). Methods A total of 206 patients were enrolled in this retrospective study. SIRT1 and VEGF-C protein expression was detected by immunohistochemical staining. Peritumoral lymphatic microvessel density (LVD) and LVI were evaluated by immunostaining for D2-40. Statistical analysis was then preformed to investigate the relevance of SIRT1 expression and various clinicopathologic features and to examine the effect of SIRT1 on tumor-induced lymphangiogenesis, LVI and prognosis. Results SIRT1 positive expression was identified in 95 cases in the nucleus and was significantly correlated with T status (P < 0.001), disease stage (P = 0.001), VEGF-C positive expression (P = 0.015), high LVD (P = 0.013) and positive LVI (P = 0.015). Patients with SIRT1 positive expression, high LVD and positive LVI had a significantly unfavorable 5-year disease free survival (P < 0.001, P = 0.030, and P < 0.001, respectively) and overall survival (P < 0.001, P = 0.017, and P < 0.001, respectively). However, based on multivariate Cox regression analysis, only SIRT1 positive expression and positive LVI were significant independent prognosticators of poor disease-free survival (P = 0.029 and 0.018, respectively) and overall survival (P = 0.045 and 0.031, respectively). Conclusions SIRT1 positive expression was significantly associated with tumor progression, lymphangiogenesis, LVI and poor survival in pN0 ESCC patients. Our research shows a utilization of SIRT1 in prognosing poor survival and providing possible target for ESCC patients through inhibiting its lymphangiogenesis activity.
Collapse
Affiliation(s)
- Guan-Qing Chen
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Hui Tian
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Wei-Ming Yue
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Lin Li
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Shu-Hai Li
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Lei Qi
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Cun Gao
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Li-Bo Si
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Ming Lu
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| | - Fei Feng
- Department of Thoracic Surgery, Qi Lu Hospital, Shandong University, Wen hua xi lu 107#, Jinan, 250012 Shandong Province China
| |
Collapse
|
114
|
de Sousa EA, Lourenço SV, de Moraes FPP, Vartanian JG, Gonçalves-Filho J, Kowalski LP, Soares FA, Coutinho-Camillo CM. Head and neck squamous cell carcinoma lymphatic spread and survival: Relevance of vascular endothelial growth factor family for tumor evaluation. Head Neck 2014; 37:1410-6. [PMID: 24824527 DOI: 10.1002/hed.23765] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/14/2014] [Accepted: 05/10/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is primarily a locoregional disease in which the cervical lymph nodes are the chief site of metastasis. The purpose of this study was to examine the relationship between lymphangiogenesis and clinicopathological aspects of HNSCC and its metastasis. METHODS Fifty-two patients with HNSCC and metastatic lymph nodes from 21 of these subjects were analyzed by immunohistochemistry. RESULTS The HNSCC samples were predominantly negative for vascular endothelial growth factor (VEGF)-C, VEGF-D, and vascular endothelial growth factor receptor (VEGFR)3. There was an association between the density of lymph vessels (measured by D2-40 staining) in the lymph nodes and advanced-stage tumors. There was no link between the expression of these proteins and survival rates. CONCLUSION Although lymphatic spread is a significant event in the progression of HNSCC, the expression of VEGF-C, VEGF-D, and VEGFR3 does not correlate with clinicopathological characteristics, suggesting that other signaling pathways mediate lymphangiogenesis in HNSCC.
Collapse
Affiliation(s)
| | | | | | - José Guilherme Vartanian
- Department of Head and Neck Surgery and Otorhinolaryngology, AC Camargo Cancer Center, São Paulo, Brazil
| | - João Gonçalves-Filho
- Department of Head and Neck Surgery and Otorhinolaryngology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaryngology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Fernando Augusto Soares
- Department of Pathology, AC Camargo Cancer Center, São Paulo, Brazil.,Department of General Pathology, Dental School, University of São Paulo, Brazil
| | | |
Collapse
|
115
|
Moens S, Goveia J, Stapor PC, Cantelmo AR, Carmeliet P. The multifaceted activity of VEGF in angiogenesis - Implications for therapy responses. Cytokine Growth Factor Rev 2014; 25:473-82. [PMID: 25169850 DOI: 10.1016/j.cytogfr.2014.07.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/15/2014] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a key growth factor driving angiogenesis (i.e. the formation of new blood vessels) in health and disease. Pharmacological blockade of VEGF signaling to inhibit tumor angiogenesis is clinically approved but the survival benefit is limited as patients invariably acquire resistance. This is partially mediated by the intrinsic flexibility of tumor cells to adapt to VEGF-blockade. However, it has become clear that tumor stromal cells also contribute to the resistance. Originally, VEGF was thought to specifically target endothelial cells (ECs) but it is now clear that many stromal cells also respond to VEGF signaling, making anti-VEGF therapy more complex than initially anticipated. A more comprehensive understanding of the complex responses of stromal cells to VEGF-blockade might inform the design of improved anti-angiogenic agents.
Collapse
Affiliation(s)
- Stijn Moens
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Peter C Stapor
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium.
| |
Collapse
|
116
|
Astin JW, Jamieson SMF, Eng TCY, Flores MV, Misa JP, Chien A, Crosier KE, Crosier PS. An in vivo antilymphatic screen in zebrafish identifies novel inhibitors of mammalian lymphangiogenesis and lymphatic-mediated metastasis. Mol Cancer Ther 2014; 13:2450-62. [PMID: 25053822 DOI: 10.1158/1535-7163.mct-14-0469-t] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The growth of new lymphatic vessels (lymphangiogenesis) in tumors is an integral step in the metastatic spread of tumor cells, first to the sentinel lymph nodes that surround the tumor and then elsewhere in the body. Currently, no selective agents designed to prevent lymphatic vessel growth have been approved for clinical use, and there is an important potential clinical niche for antilymphangiogenic agents. Using a zebrafish phenotype-based chemical screen, we have identified drug compounds, previously approved for human use, that have antilymphatic activity. These include kaempferol, a natural product found in plants; leflunomide, an inhibitor of pyrimidine biosynthesis; and cinnarizine and flunarizine, members of the type IV class of calcium channel antagonists. Antilymphatic activity was confirmed in a murine in vivo lymphangiogenesis Matrigel plug assay, in which kaempferol, leflunomide, and flunarizine prevented lymphatic growth. We show that kaempferol is a novel inhibitor of VEGFR2/3 kinase activity and is able to reduce the density of tumor-associated lymphatic vessels as well as the incidence of lymph node metastases in a metastatic breast cancer xenograft model. However, in this model, kaempferol administration was also associated with tumor deposits in the pancreas and diaphragm, and flunarizine was found to be tumorigenic. Although this screen revealed that zebrafish is a viable platform for the identification and development of mammalian antilymphatic compounds, it also highlights the need for focused secondary screens to ensure appropriate efficacy of hits in a tumor context.
Collapse
Affiliation(s)
- Jonathan W Astin
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand. Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Tiffany C Y Eng
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand. Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Maria V Flores
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - June P Misa
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Annie Chien
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn E Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand. Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand. Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
117
|
Xu GC, Zhang P, Leng F, Pan L, Li ZY, Yu DD, Shan Y, Yuan QZ, Wen Y, Mu B, Shi HS, Chen X, Wang CT. Inhibition of lymphatic metastases by a survivin dominant-negative mutant. Oncol Res 2014; 20:579-87. [PMID: 24139416 DOI: 10.3727/096504013x13775486749416] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Metastasis is the most lethal attribute of human malignancy. High-level expression of survivin is involved in both carcinogenesis and angiogenesis in cancer. Previous studies indicate that a mutation of the threonine residue at position 34 (Thr34Ala) of survivin generates a dominant-negative mutant that induces apoptosis, inhibits angiogenesis, and suppresses highly metastatic breast carcinoma in mouse models. We investigated the efficacy of gene therapy with a survivin dominant-negative mutant and possible factors related to lymph node metastasis. The metastasis rate was compared between each group in order to find a survivin-targeted therapy against lymphangiogenesis in its earliest stages. We established lymph node metastasis models and treated animals with H22 tumors with Lip-mSurvivinT34A (Lip-mS), Lip-plasmid (Lip-P), or normal saline (NS). Eight days after the last dose, five randomly chosen mice from each group were sacrificed. We detected the apoptotic index, microvessel density (MVD), lymphatic microvessel density (LMVD), and the expression of VEGF-D with immunohistochemistry. After the remaining animals were sacrificed, we compared the tumor-infiltrated lymph nodes in each group. Administration of mSurvivinT34A plasmid complexed with cationic liposome (DOTAP/chol) resulted in the efficacious inhibition of tumor growth and lymph node metastasis within the mouse H22 tumor model. These responses were associated with tumor cell apoptosis, and angiogenesis and lymphangiogenesis inhibition. Our results suggested that Lip-mSurvivinT34A induced apoptosis and inhibited tumor angiogenesis and lymphangiogenesis, thus suppressing tumor growth and lymphatic metastasis. The mSurvivinT34A survivin mutant is a promising strategy of gene therapy to inhibit lymphatic metastasis.
Collapse
Affiliation(s)
- Guang-Chao Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
The main function of the lymphatic system is to control and maintain fluid homeostasis, lipid transport, and immune cell trafficking. In recent years, the pathological roles of lymphangiogenesis, the generation of new lymphatic vessels from preexisting ones, in inflammatory diseases and cancer progression are beginning to be elucidated. Sphingosine-1-phosphate (S1P), a bioactive lipid, mediates multiple cellular events, such as cell proliferation, differentiation, and trafficking, and is now known as an important mediator of inflammation and cancer. In this review, we will discuss recent findings showing the emerging role of S1P in lymphangiogenesis, in inflammation, and in cancer.
Collapse
|
119
|
Liu J, Liu C, Qiu L, Li J, Zhang P, Sun Y. Overexpression of both platelet-derived growth factor-BB and vascular endothelial growth factor-C and its association with lymphangiogenesis in primary human non-small cell lung cancer. Diagn Pathol 2014; 9:128. [PMID: 24972450 PMCID: PMC4085714 DOI: 10.1186/1746-1596-9-128] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/13/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Metastatic spread of tumor through lymphatic vasculature is an important adverse prognostic factor in a variety of human cancer and tumor lymphangiogenesis requires the interplay of several growth factors. Platelet-derived growth factor (PDGF)-BB and vascular endothelial growth factor (VEGF)-C are two important molecules involving in tumor metastasis and lymphangiogenesis. Therefore, the aim of this study was to investigate the coexpression of PDGF-BB and VEGF-C in primary human non-small cell lung cancer (NSCLC) and its association with lymphangiogenesis. METHODS Using immunohistochemical staining, PDGF-BB and VEGF-C expression were detected in 109 primary NSCLC tissues, while the lymphatic micro-vessel density (LMVD) was counted. RESULTS Of 109 cases, PDGF-BB and VEGF-C overexpression was 66.97% (73/109) and 65.14% (71/109), respectively. 52 (47.7%) had overexpression of both PDGF-BB and VEGF-C (P+V+), 21 (19.3%) overexpression of PDGF-BB but low expression of VEGF-C (P+V-), 19(17.4%) overexpression of VEGF-C but low expression of PDGF-BB (P-V+) and 17(15.6%) low expression of both PDGF-BB and VEGF-C (P-V-). PDGF-BB expression was positively related to that of VEGF-C (r=0.451, p=0.034). LMVD in cases with P+V+was much higher than those with P-V- (p=0.004). In addition, the patients with P+V+were younger and also had larger tumor size, more likely lymph node metastasis and worse histological differentiation than those with P-V-. Moreover, the overall survival (OS) of patients with P+V+was shorter than those with P-V- (p=0.015). CONCLUSION Coexpression of both PDGF-BB and VEGF-C was associated with lymphangiogenesis and poor prognosis in NSCLC, and might play a critical role in NSCLC progression. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/2261801312571320.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Affiliated to Shandong University, No, 105,Jiefang Road, Jinan, Shandong 250013, P,R, China.
| |
Collapse
|
120
|
Van't Hull EF, Bron S, Henry L, Ifticene-Treboux A, Turrini R, Coukos G, Delaloye JF, Doucey MA. Bone marrow-derived cells are implicated as a source of lymphatic endothelial progenitors in human breast cancer. Oncoimmunology 2014; 3:e29080. [PMID: 25101222 PMCID: PMC4121340 DOI: 10.4161/onci.29080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/23/2014] [Accepted: 04/30/2014] [Indexed: 12/31/2022] Open
Abstract
Bone marrow-derived endothelial progenitor cells (EPCs) infiltrate into sites of neovascularization in adult tissues and mature into functional blood endothelial cells (BECs) during a process called vasculogenesis. Human marrow-derived EPCs have recently been reported to display a mixed myeloid and lymphatic endothelial cell (LEC) phenotype during inflammation-induced angiogenesis; however, their role in cancer remains poorly understood. We report the in vitro differentiation of human cord blood CD133+CD34+ progenitors into podoplanin+ cells expressing both myeloid markers (CD11b, CD14) and the canonical LEC markers vascular endothelium growth factor receptor 3 (VEGFR-3), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1), and prospero homeobox 1 (PROX-1). These podoplanin+ cells displayed sprouting behavior comparable to that of LECs in vitro and a dual hemangiogenic and lymphangiogenic activity in vivo in an endothelial cell sprouting assay and corneal vascularization assay, respectively. Furthermore, these cells expressed vascular endothelium growth factor (VEGF) family members A, -C, and -D. Thus, bone-marrow derived EPCs stimulate hemangiogenesis and lymphangiogenesis through their ability to differentiate into LECs and to produce angiogenic factors. Importantly, plasma from patients with breast cancer induced differentiation of CD34+ cord blood progenitors into hemangiogenic and lymphangiogenic CD11b+ myeloid cells, whereas plasma from healthy women did not have this effect. Consistent with these findings, circulating CD11b+ cells from breast cancer patients, but not from healthy women, displayed a similar dual angiogenic activity. Taken together, our results show that marrow-derived EPCs become hemangiogenic and lymphangiogenic upon exposure to cancer plasma. These newly identified functions of bone-marrow derived EPCs are expected to influence the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
| | - Sylvian Bron
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | - Luc Henry
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | | | - Riccardo Turrini
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | | | - Marie-Agnès Doucey
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| |
Collapse
|
121
|
Identification of genes regulating migration and invasion using a new model of metastatic prostate cancer. BMC Cancer 2014; 14:387. [PMID: 24885350 PMCID: PMC4046438 DOI: 10.1186/1471-2407-14-387] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 05/22/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Understanding the complex, multistep process of metastasis remains a major challenge in cancer research. Metastasis models can reveal insights in tumor development and progression and provide tools to test new intervention strategies. METHODS To develop a new cancer metastasis model, we used DU145 human prostate cancer cells and performed repeated rounds of orthotopic prostate injection and selection of subsequent lymph node metastases. Tumor growth, metastasis, cell migration and invasion were analyzed. Microarray analysis was used to identify cell migration- and cancer-related genes correlating with metastasis. Selected genes were silenced using siRNA, and their roles in cell migration and invasion were determined in transwell migration and Matrigel invasion assays. RESULTS Our in vivo cycling strategy created cell lines with dramatically increased tumorigenesis and increased ability to colonize lymph nodes (DU145LN1-LN4). Prostate tumor xenografts displayed increased vascularization, enlarged podoplanin-positive lymphatic vessels and invasive margins. Microarray analysis revealed gene expression profiles that correlated with metastatic potential. Using gene network analysis we selected 3 significantly upregulated cell movement and cancer related genes for further analysis: EPCAM (epithelial cell adhesion molecule), ITGB4 (integrin β4) and PLAU (urokinase-type plasminogen activator (uPA)). These genes all showed increased protein expression in the more metastatic DU145-LN4 cells compared to the parental DU145. SiRNA knockdown of EpCAM, integrin-β4 or uPA all significantly reduced cell migration in DU145-LN4 cells. In contrast, only uPA siRNA inhibited cell invasion into Matrigel. This role of uPA in cell invasion was confirmed using the uPA inhibitors, amiloride and UK122. CONCLUSIONS Our approach has identified genes required for the migration and invasion of metastatic tumor cells, and we propose that our new in vivo model system will be a powerful tool to interrogate the metastatic cascade in prostate cancer.
Collapse
|
122
|
Riquet M, Legras A, Mordant P, Rivera C, Arame A, Gibault L, Foucault C, Dujon A, Le Pimpec Barthes F. Number of mediastinal lymph nodes in non-small cell lung cancer: a Gaussian curve, not a prognostic factor. Ann Thorac Surg 2014; 98:224-31. [PMID: 24820386 DOI: 10.1016/j.athoracsur.2014.03.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/11/2014] [Accepted: 03/20/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND It has been proposed that examining a greater number of lymph nodes (LNs) in patients with non-small-cell lung cancer (NSCLC) treated by surgical resection may increase the likelihood of proper staging and affect outcome. Our purpose was to evaluate the interindividual variability and prognostic relevance of the number of LNs harvested during complete pulmonary and mediastinal lymphadenectomy performed for NSCLC. METHODS We prospectively collected and retrospectively reviewed the data from 1,095 patients who underwent lung cancer resection in association with systematic lymphadenectomy and pulmonary and mediastinal LN counts from 2004 to 2009. We analyzed the interindividual variability and prognostic impact of the number of LNs on overall survival (OS). RESULTS The mean number of harvested pulmonary and mediastinal LNs was 17.4±7.3 (range, 1-65) and was higher in male patients, right lung surgical procedures, lobectomy and pneumonectomy, N2 disease, and pIII stage. The mean number of harvested mediastinal LNs was 10.7±5.6 and was normally distributed (range, 0-49; median, 10). The 5-year survival rate was 53.8%. Overall survival was influenced by the number of involved stations (single-station versus multi-station disease, 5-year survival rates 31.5% versus 16.9%, respectively; p=0.041) but not by the number of harvested LNs, the number of harvested mediastinal LNs, or the number of positive mediastinal LNs. CONCLUSIONS After lung cancer resection and complete lymphadenectomy, the number of LNs is subject to normally distributed interindividual variability, with no significant impact on OS. Recommending an optimal number of nodes is therefore arbitrary. Instead, our recommendation is to perform a complete systematic pulmonary and mediastinal lymphadenectomy following established anatomical boundaries.
Collapse
Affiliation(s)
- Marc Riquet
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France.
| | - Antoine Legras
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Pierre Mordant
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Caroline Rivera
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Alex Arame
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Laure Gibault
- Department of Pathology, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Christophe Foucault
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Antoine Dujon
- Department of General Thoracic Surgery, Cedar Surgical Centre, Bois-Guillaume, France
| | - Françoise Le Pimpec Barthes
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| |
Collapse
|
123
|
Liu H, Yang Y, Xiao J, Yang S, Liu Y, Kang W, Li X, Zhang F. Semaphorin 4D expression is associated with a poor clinical outcome in cervical cancer patients. Microvasc Res 2014; 93:1-8. [PMID: 24603190 DOI: 10.1016/j.mvr.2014.02.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 01/13/2023]
Abstract
Lymphangiogenesis is thought to be essential for cancer progression, making it an important target in cancer therapy. Lymphangiogenic factors (VEGF-C and VEGF-D) are upregulated in various tumors/cancers, and play an important role in lymphangiogenesis and lymph node metastasis. Similarly, semaphorin 4D (Sema4D) is a potent inducer of angiogenesis, and its overexpression is associated with tumor progression and poor prognosis in a variety of malignancies. However, little is known regarding the functional relationship between Sema4D and VEGF-C/VEGF-D and in the mediation of lymphangiogenesis and lymph node metastasis and clinical outcome. The current study aimed to evaluate the effect of Sema4D expression on outcome in patients with cervical cancer, and to explore the molecular mechanism of Sema4D in tumor progression. We evaluated Sema4D expression, density of lymphatic vessels, and invasion of lymphatic vessels with immunohistochemical methods in 232 human cervical cancers with long-term follow-up. Sema4D expression was correlated with patho-clinical parameters and patients' outcome. A cervical cancer cell line was used to investigate the contribution of sema4D to tumor progression by studying the role of Sema4D in VEGF-C/-D and cell migration using reverse transcription-polymerase chain reaction and Western blotting. We observed that Sema4D expression was higher in metastatic cervical cancer than in nonmetastatic cervical cancer (P<0.001). CD34-positive or D2-40-positive lymphatic vessel density was significantly increased in cases with lymph node metastasis compared with those without lymph node metastasis. The increased Sema4D expression was associated with VEGF-C/-D, the presence of lymphatic invasion, the occurrence of lymph node metastasis, and FIGO stage. We also observed a novel association between Sema4D upregulation and poor prognosis in cervical cancer. In vitro, the Sema4D inhibitory antibody and Sema4D-shRNA suppressed VEGF-C and VEGF-D in the human cervical carcinoma cell lines HeLa, Siha, and Caski cells. Invasiveness assay demonstrated that Sema4D could augment the invasive potential of the tumor cells in the cervical cancer lines and induction of cellular invasiveness by Sema4D stimulation could be inhibited by knockdown of plexinB1 by siRNA. Further mechanistic investigations of tumor cell invasiveness showed that Sema4D could induce activation of GTPase Ras homolog gene family, member A (RhoA), MAPK and AKT. In addition, plexinB1 knockdown by siRNA could suppress the Sema4D signal transmitted to MAPK and Akt. Taken together, these results suggest that Sema4D autocrine within tumor cells contributes to enhanced invasion and tumor progression through increased motility of cervical cancer and VEGF-C/-D-mediated lymphangiogenesis. Sema4D might be useful as a molecular marker of poor prognosis in cervical cancer.
Collapse
Affiliation(s)
- Huidong Liu
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yanmei Yang
- Cancer Institute, Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Jianbing Xiao
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Shucai Yang
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yan Liu
- Department of General Surgery, Mudanjiang First Hospital, 4 Jianwei Road, 157011 Mudanjiang, China
| | - Wenhui Kang
- Department of Radiotherapy, The Third Affiliated Hospital of Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Xinlei Li
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Fengmin Zhang
- Department of Medical Microbiology, Heilongjiang Province Key Laboratory for Immunity and Infection, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China.
| |
Collapse
|
124
|
Abstract
Malignant tumors release growth factors such as VEGF-C to induce lymphatic vessel expansion (lymphangiogenesis) in primary tumors and in draining sentinel LNs, thereby promoting LN metastasis. Surprising recent evidence suggests that lymphatic vessels do not merely represent passive channels for tumor spread, but that they may actively promote tumor cell recruitment to LNs, cancer stem cell survival, and immune modulation. New imaging approaches allow the sensitive visualization of the earliest LN metastases and the quantitative, noninvasive measurement of the function of tumor-draining lymphatic vessels, with potential applications in the development of biomarkers for prognosis and measurement of therapeutic response.
Collapse
|
125
|
Legras A, Mordant P, Arame A, Foucault C, Dujon A, Le Pimpec Barthes F, Riquet M. Long-term survival of patients with pN2 lung cancer according to the pattern of lymphatic spread. Ann Thorac Surg 2014; 97:1156-62. [PMID: 24582052 DOI: 10.1016/j.athoracsur.2013.12.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/13/2013] [Accepted: 12/30/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND N2 involvement has dramatic consequences on the prognosis and management of patients with non-small cell lung cancer (NSCLC). N2-NSCLC may present with or without N1 involvement, constituting non-skip (pN1N2) and skip (pN0N2) diseases, respectively. As the prognostic impact of this subclassification is still a matter of debate, we analyzed the prognosis of pN2 patients according to the pN1-involvement and the number of N2-stations concerned. METHODS The medical records of consecutive patients who underwent surgery for pN2-NSCLC in 2 French centers between 1980 and 2009 were prospectively collected and retrospectively reviewed. Patients undergoing induction therapy, exploratory thoracotomy, incomplete mediastinal lymphadenectomy, or incomplete resections were excluded. The prognoses of pN1N2 and pN0N2 patients were first compared, and then deciphered according to the number of N2 stations involved (single-station: 1S, multi-station: 2S). RESULTS All together, 871 patients underwent first-line complete surgical resection for pN2-NSCLC during the study period, including 258 pN0N2 (29.6%) and 613 pN1N2 (70.4%) patients. Mean follow-up was 72.8±48 months. Median, 5- and 10-year survivals were, respectively, 30 months, 34%, and 24% for pN0N2 and 20 months, 21%, and 14% for pN1N2 patients (p<0.001). Multivariate analysis revealed 3 different prognostic groups; ie, favorable in pN0N2-1S disease, intermediate in pN0N2-2S and pN1N2-1S diseases, and poor in pN1N2-2S disease (p<0.001). CONCLUSIONS Among pN2 patients, the combination of N1 involvement (pN0N2 vs pN1N2) and number of involved N2 stations (1S vs 2S) are independent prognostic factors. These results might be taken into consideration to sub-classify the heterogeneous pN2-NSCLC group of patients.
Collapse
Affiliation(s)
- Antoine Legras
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Pierre Mordant
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Alex Arame
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Christophe Foucault
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | | | - Françoise Le Pimpec Barthes
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France
| | - Marc Riquet
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Descartes University, Paris, France.
| |
Collapse
|
126
|
Du LC, Chen XC, Wang D, Wen YJ, Wang CT, Wang XM, Kan B, Wei YQ, Zhao X. VEGF-D-induced draining lymphatic enlargement and tumor lymphangiogenesis promote lymph node metastasis in a xenograft model of ovarian carcinoma. Reprod Biol Endocrinol 2014; 12:14. [PMID: 24502459 PMCID: PMC3929486 DOI: 10.1186/1477-7827-12-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 12/29/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)-D has been shown to promote lymph node metastasis in several cancers. Although generally overexpressed in ovarian carcinoma, its role in nodal dissemination of this cancer is unclear. To clarify the role of VEGF-D and the underlying molecular mechanisms, we investigated the function of VEGF-D using a mouse xenograft model of ovarian cancer. METHODS Human ovarian serous adenocarcinoma SKOV3 cells were transfected with VEGF-D recombinant plasmid DNA, or with control vectors. The cells were injected subcutaneously into the footpads of nude mice. Tumor growth was evaluated weekly. Draining lymphatics were observed grossly with Evan's blue lymphangiography. Tumoral lymphatics were delineated with both Evan's blue and LYVE-1 immunostaining. Tumor metastases to lymph nodes were evaluated by H&E and CA125/CD40 staining. Expression of VEGF-D in primary tumors and levels of CA125 in involved lymph nodes were examined by immunohistochemistry. Tumor cell apoptosis was analyzed by Hoechst dyeing. RESULTS Mice bearing VEGF-D overexpressing xenografts showed a significantly higher rate of lymph node metastasis and markedly greater tumor volume compared with the controls. The functional lymphatic vessels were denser and enlarged in marginal and central tumor portions. Additionally, higher CA125 expression was observed in the involved lymph nodes. Mice bearing VEGF-D overexpressing xenografts also exhibited a markedly lower apoptotic index compared with the controls. CONCLUSIONS Our data demonstrate the important role of VEGF-D in promoting lymph node metastasis by increasing tumor lymphangiogenesis, stimulating draining lymphatic vessel formation, and enhancing tumor invasiveness. Our findings show that VEGF-D can be a promising therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Li-Cheng Du
- Department of Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jingwu Road, Jinan, China
| | - Xian-Cheng Chen
- Department of Gynecology and Obstetrics, Second West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Dong Wang
- Department of Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jingwu Road, Jinan, China
| | - Yan-Jun Wen
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Chun-Ting Wang
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Xue-Mei Wang
- Department of Gynecology and Obstetrics, Second West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Bing Kan
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Yu-Quan Wei
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Second West China Hospital, West China Medical School, Sichuan University, South Renmin Road, Chengdu, China
| |
Collapse
|
127
|
Cazes A, Gibault L, Rivera C, Mordant P, Riquet M. [Lymphatic extension and lymphangiogenesis in non-small cell lung cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2014; 70:26-31. [PMID: 24566036 DOI: 10.1016/j.pneumo.2013.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/23/2013] [Indexed: 06/03/2023]
Abstract
Lymph node metastasis is a major adverse prognostic factor of malignant tumors, including non-small cell lung carcinoma (NSCLC). However the characterization of tumor associated lymphatic vessels and lymphangiogenic mediators in NSCLC are recent and their prognostic role is debated. Lymphatic vascular invasion (LVI) appears like a robust adverse prognostic factor when reported in NSCLC. This parameter should be better standardized and could be of use in adjuvant therapy indications. Moreover, anti-lymphangiogenesis therapies are currently under investigation and may become part of the anti-cancer strategy.
Collapse
Affiliation(s)
- A Cazes
- Service d'anatomie pathologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - L Gibault
- Service d'anatomie pathologique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - C Rivera
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - P Mordant
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France
| | - M Riquet
- Service de chirurgie thoracique, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France; Unversité Paris Descartes, 12, rue de l'École-de-Médecine, 75006 Paris, France.
| |
Collapse
|
128
|
Farina AR, Mackay AR. Gelatinase B/MMP-9 in Tumour Pathogenesis and Progression. Cancers (Basel) 2014; 6:240-96. [PMID: 24473089 PMCID: PMC3980597 DOI: 10.3390/cancers6010240] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 12/14/2022] Open
Abstract
Since its original identification as a leukocyte gelatinase/type V collagenase and tumour type IV collagenase, gelatinase B/matrix metalloproteinase (MMP)-9 is now recognised as playing a central role in many aspects of tumour progression. In this review, we relate current concepts concerning the many ways in which gelatinase B/MMP-9 influences tumour biology. Following a brief outline of the gelatinase B/MMP-9 gene and protein, we analyse the role(s) of gelatinase B/MMP-9 in different phases of the tumorigenic process, and compare the importance of gelatinase B/MMP-9 source in the carcinogenic process. What becomes apparent is the importance of inflammatory cell-derived gelatinase B/MMP-9 in tumour promotion, early progression and triggering of the "angiogenic switch", the integral relationship between inflammatory, stromal and tumour components with respect to gelatinase B/MMP-9 production and activation, and the fundamental role for gelatinase B/MMP-9 in the formation and maintenance of tumour stem cell and metastatic niches. It is also apparent that gelatinase B/MMP-9 plays important tumour suppressing functions, producing endogenous angiogenesis inhibitors, promoting inflammatory anti-tumour activity, and inducing apoptosis. The fundamental roles of gelatinase B/MMP-9 in cancer biology underpins the need for specific therapeutic inhibitors of gelatinase B/MMP-9 function, the use of which must take into account and substitute for tumour-suppressing gelatinase B/MMP-9 activity and also limit inhibition of physiological gelatinase B/MMP-9 function.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L'Aquila 67100, Italy.
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L'Aquila 67100, Italy.
| |
Collapse
|
129
|
Morfoisse F, Kuchnio A, Frainay C, Gomez-Brouchet A, Delisle MB, Marzi S, Helfer AC, Hantelys F, Pujol F, Guillermet-Guibert J, Bousquet C, Dewerchin M, Pyronnet S, Prats AC, Carmeliet P, Garmy-Susini B. Hypoxia induces VEGF-C expression in metastatic tumor cells via a HIF-1α-independent translation-mediated mechanism. Cell Rep 2014; 6:155-67. [PMID: 24388748 DOI: 10.1016/j.celrep.2013.12.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 10/28/2013] [Accepted: 12/06/2013] [Indexed: 01/04/2023] Open
Abstract
Various tumors metastasize via lymph vessels and lymph nodes to distant organs. Even though tumors are hypoxic, the mechanisms of how hypoxia regulates lymphangiogenesis remain poorly characterized. Here, we show that hypoxia reduced vascular endothelial growth factor C (VEGF-C) transcription and cap-dependent translation via the upregulation of hypophosphorylated 4E-binding protein 1 (4E-BP1). However, initiation of VEGF-C translation was induced by hypoxia through an internal ribosome entry site (IRES)-dependent mechanism. IRES-dependent VEGF-C translation was independent of hypoxia-inducible factor 1α (HIF-1α) signaling. Notably, the VEGF-C IRES activity was higher in metastasizing tumor cells in lymph nodes than in primary tumors, most likely because lymph vessels in these lymph nodes were severely hypoxic. Overall, this transcription-independent but translation-dependent upregulation of VEGF-C in hypoxia stimulates lymphangiogenesis in tumors and lymph nodes and may contribute to lymphatic metastasis.
Collapse
Affiliation(s)
- Florent Morfoisse
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Anna Kuchnio
- Vesalius Research Center, VIB, University of Leuven, 3000 Leuven, Belgium
| | - Clement Frainay
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Anne Gomez-Brouchet
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Marie-Bernadette Delisle
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Stefano Marzi
- UPR 9002 CNRS-ARN, Université De Strasbourg, IBMC, 67084 Strasbourg, France
| | | | - Fransky Hantelys
- Université de Toulouse, UPS, TRADGENE, EA4554, 31432 Toulouse, France
| | - Francoise Pujol
- Université de Toulouse, UPS, TRADGENE, EA4554, 31432 Toulouse, France
| | - Julie Guillermet-Guibert
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Corinne Bousquet
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | - Mieke Dewerchin
- Vesalius Research Center, VIB, University of Leuven, 3000 Leuven, Belgium
| | - Stephane Pyronnet
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France
| | | | - Peter Carmeliet
- Vesalius Research Center, VIB, University of Leuven, 3000 Leuven, Belgium
| | - Barbara Garmy-Susini
- Inserm, U1037, 31432 Toulouse, France; Université de Toulouse, UPS, Cancer Research Center of Toulouse, Equipe Labellisee Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer, 31432 Toulouse, France.
| |
Collapse
|
130
|
Moreira Â, Pereira SS, Machado CL, Morais T, Costa M, Monteiro MP. Obesity inhibits lymphangiogenesis in prostate tumors. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 7:348-352. [PMID: 24427356 PMCID: PMC3885490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/12/2013] [Indexed: 06/03/2023]
Abstract
Lymphangiogenesis is the process that leads to new lymphatic vessels formation from preexisting blood vessels in the presence of appropriate inducing signals, which in pathologic conditions such as cancer, may contribute to tumor cells dissemination. The aim of the present study was to study the role of obesity, leptin and insulin in tumor lymphangiogenesis. For that, we have quantified the lymphatic vessels in prostate tumors through their immunohistochemistry staining by Lyve-1 in RM1 prostate tumors induced in different obese mice models (ob/ob, db/db and diet induced obese (DIO) and in normal weight C57BL/6J mice (control). Lymph vessels density was determined by Lyve-1 immunohistochemistry of prostate adenocarcinomas, while the percentage of the Lyve-1 stained area and lymphatic vessels number were obtained using a morphometric computerized tool. Obese ob/ob and DIO mice presented prostate tumors that were significantly larger (p<0.001) than controls, while tumors of db/db mice were significantly smaller (p=0.047). Lyve-1 expression was significantly higher in prostate tumors of DIO mice compared to tumors of db/db mice (p<0.05); furthermore Lyve-1 expression was negatively correlated with the percentage of the epididymal fat and body weight (p<0.01). No significantly correlations were found between Lyve-1 expression and tumor weight and leptin or insulin plasma levels. Our results suggest that obesity may have a protective effect against prostate cancer dissemination by inhibiting lymphangiogenesis through a still unidentified mechanism that appears not to involve leptin or insulin.
Collapse
Affiliation(s)
- Ângela Moreira
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| | - Sofia S Pereira
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| | - Christiane L Machado
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| | - Tiago Morais
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| | - Madalena Costa
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| | - Mariana P Monteiro
- Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Institute for Biomedical Sciences Abel Salazar (ICBAS), University of Porto Portugal
| |
Collapse
|
131
|
Li D, Xie K, Ding G, Li J, Chen K, Li H, Qian J, Jiang C, Fang J. Tumor resistance to anti-VEGF therapy through up-regulation of VEGF-C expression. Cancer Lett 2013; 346:45-52. [PMID: 24333721 DOI: 10.1016/j.canlet.2013.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/14/2013] [Accepted: 12/04/2013] [Indexed: 12/22/2022]
Abstract
Increasing evidence has indicated that prolonged use of anti-VEGF (vascular endothelial growth factor) agents for cancer therapy promotes tumor resistance. To gain insight into the molecular mechanism underlying resistance to anti-VEGF therapy, we developed a mouse Lewis lung carcinoma (LLC) cell line that is resistant to treatment with a potent VEGF inhibitor, VEGF-Trap, through repeated in vivo selection. We compared the transcriptome profiles of resistant and non-resistant tumor cells using RNA-seq analysis. VEGF-C was significantly up-regulated in resistant tumor cells, as determined by quantitative real-time PCR and immunohistochemical analyses. Inhibition of VEGF-C in resistant cells suppressed endothelial cell migration in vitro and partially restored sensitivity to VEGF-Trap treatment in vivo. Our findings indicate that tumors may develop resistance to anti-VEGF therapy by activating the VEGF-C pathway.
Collapse
Affiliation(s)
- Dong Li
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kun Xie
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Guitao Ding
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jie Li
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kaiming Chen
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hongwen Li
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jie Qian
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Cizhong Jiang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jianmin Fang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
132
|
Valtcheva N, Primorac A, Jurisic G, Hollmén M, Detmar M. The orphan adhesion G protein-coupled receptor GPR97 regulates migration of lymphatic endothelial cells via the small GTPases RhoA and Cdc42. J Biol Chem 2013; 288:35736-48. [PMID: 24178298 DOI: 10.1074/jbc.m113.512954] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The important role of the lymphatic vascular system in pathological conditions such as inflammation and cancer has been increasingly recognized, but its potential as a pharmacological target is poorly exploited. Our study aimed at the identification and molecular characterization of lymphatic-specific G protein-coupled receptors (GPCRs) to assess new targets for pharmacological manipulation of the lymphatic vascular system. We used a TaqMan quantitative RT-PCR-based low density array to determine the GPCR expression profiles of ex vivo isolated intestinal mouse lymphatic (LECs) and blood vascular endothelial cells (BECs). GPR97, an orphan adhesion GPCR of unknown function, was the most highly and specifically expressed GPCR in mouse lymphatic endothelium. Using siRNA silencing, we found that GPR97-deficient primary human LECs displayed increased adhesion and collective cell migration, whereas single cell migration was decreased as compared with nontargeting siRNA-transfected control LECs. Loss of GPR97 shifted the ratio of active Cdc42 and RhoA and initiated cytoskeletal rearrangements, including F-actin redistribution, paxillin and PAK4 phosphorylation, and β1-integrin activation. Our data suggest a possible role of GPR97 in lymphatic remodeling and furthermore provide the first insights into the biological functions of GPR97.
Collapse
Affiliation(s)
- Nadejda Valtcheva
- From the Institute of Pharmaceutical Sciences, ETH Zurich, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
133
|
Zorgetto VA, Silveira GG, Oliveira-Costa JP, Soave DF, Soares FA, Ribeiro-Silva A. The relationship between lymphatic vascular density and vascular endothelial growth factor A (VEGF-A) expression with clinical-pathological features and survival in pancreatic adenocarcinomas. Diagn Pathol 2013; 8:170. [PMID: 24138811 PMCID: PMC3816792 DOI: 10.1186/1746-1596-8-170] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/07/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a rare tumor with an extremely low survival rate. Its known risk factors include the chronic use of tobacco and excessive alcohol consumption and the presence of chronic inflammatory diseases, such as pancreatitis and type 2 diabetes. Angiogenesis and lymphangiogenesis, which have been the focus of recent research, are considered prognostic factors for cancer development. Knowing the angiogenic and lymphangiogenic profiles of a tumor may provide new insights for designing treatments according to the different properties of the tumor. The aim of this study was to evaluate the density of blood and lymphatic vessels, and the expression of VEGF-A, in pancreatic adenocarcinomas, as well as the relationship between blood and lymphatic vascular density and the prognostically important clinical-pathological features of pancreatic tumors. METHODS Paraffin blocks containing tumor samples from 100 patients who were diagnosed with pancreatic cancer between 1990 and 2010 were used to construct a tissue microarray. VEGF expression was assessed in these samples by immunohistochemistry. To assess the lymphatic and vascular properties of the tumors, 63 cases that contained sufficient material were sectioned routinely. The sections were then stained with the D2-40 antibody to identify the lymphatic vessels and with a CD34 antibody to identify the blood vessels. The vessels were counted individually with the Leica Application Suite v4 program. All statistical analyses were performed using SPSS 18.0 (Chicago, IL, USA) software, and p values ≤ 0.05 were considered significant. RESULTS In the Cox regression analysis, advanced age (p=0.03) and a history of type 2 diabetes (p=0.014) or chronic pancreatitis (p=0.02) were shown to be prognostic factors for pancreatic cancer. Blood vessel density (BVD) had no relationship with clinical-pathological features or death. Lymphatic vessel density (LVD) was inversely correlated with death (p=0.002), and by Kaplan-Meyer survival analysis, we found a significant association between low LVD (p=0.021), VEGF expression (p=0.023) and low patient survival. CONCLUSIONS Pancreatic carcinogenesis is related to a history of chronic inflammatory processes, such as type 2 diabetes and chronic pancreatitis. In pancreatic cancer development, lymphangiogenesis can be considered an early event that enables the dissemination of metastases. VEGF expression and low LVD can be considered as poor prognostic factors as tumors with this profile are fast growing and highly aggressive. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/5113892881028514.
Collapse
Affiliation(s)
| | | | | | | | | | - Alfredo Ribeiro-Silva
- Departament of Pathology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
134
|
Cao Z, Shang B, Zhang G, Miele L, Sarkar FH, Wang Z, Zhou Q. Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis. Biochim Biophys Acta Rev Cancer 2013; 1836:273-86. [PMID: 23933263 DOI: 10.1016/j.bbcan.2013.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/22/2013] [Accepted: 08/01/2013] [Indexed: 12/12/2022]
Abstract
Robust neovascularization and lymphangiogenesis have been found in a variety of aggressive and metastatic tumors. Endothelial sprouting angiogenesis is generally considered to be the major mechanism by which new vasculature forms in tumors. However, increasing evidence shows that tumor vasculature is not solely composed of endothelial cells (ECs). Some tumor cells acquire processes similar to embryonic vasculogenesis and produce new vasculature through vasculogenic mimicry, trans-differentiation of tumor cells into tumor ECs, and tumor cell-EC vascular co-option. In addition, tumor cells secrete various vasculogenic factors that induce sprouting angiogenesis and lymphangiogenesis. Vasculogenic tumor cells actively participate in the formation of vascular cancer stem cell niche and a premetastatic niche. Therefore, tumor cell-mediated neovascularization and lymphangiogenesis are closely associated with tumor progression, cancer metastasis, and poor prognosis. Vasculogenic tumor cells have emerged as key players in tumor neovascularization and lymphangiogenesis and play pivotal roles in tumor progression and cancer metastasis. However, the mechanisms underlying tumor cell-mediated vascularity as they relate to tumor progression and cancer metastasis remain unclear. Increasing data have shown that various intrinsic and extrinsic factors activate oncogenes and vasculogenic genes, enhance vasculogenic signaling pathways, and trigger tumor neovascularization and lymphangiogenesis. Collectively, tumor cells are the instigators of neovascularization. Therefore, targeting vasculogenic tumor cells, genes, and signaling pathways will open new avenues for anti-tumor vasculogenic and metastatic drug discovery. Dual targeting of endothelial sprouting angiogenesis and tumor cell-mediated neovascularization and lymphangiogenesis may overcome current clinical problems with anti-angiogenic therapy, resulting in significantly improved anti-angiogenesis and anti-cancer therapies.
Collapse
Affiliation(s)
- Zhifei Cao
- Cyrus Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | |
Collapse
|
135
|
Martinez-Corral I, Makinen T. Regulation of lymphatic vascular morphogenesis: Implications for pathological (tumor) lymphangiogenesis. Exp Cell Res 2013; 319:1618-25. [DOI: 10.1016/j.yexcr.2013.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/26/2013] [Indexed: 11/24/2022]
|
136
|
Cermenati S, Moleri S, Neyt C, Bresciani E, Carra S, Grassini DR, Omini A, Goi M, Cotelli F, François M, Hogan BM, Beltrame M. Sox18 Genetically Interacts With VegfC to Regulate Lymphangiogenesis in Zebrafish. Arterioscler Thromb Vasc Biol 2013; 33:1238-47. [DOI: 10.1161/atvbaha.112.300254] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Solei Cermenati
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Silvia Moleri
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Christine Neyt
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Erica Bresciani
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Silvia Carra
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Daniela R. Grassini
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Alice Omini
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Michela Goi
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Franco Cotelli
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Mathias François
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Benjamin M. Hogan
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Monica Beltrame
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| |
Collapse
|
137
|
Pasquali S, van der Ploeg APT, Mocellin S, Stretch JR, Thompson JF, Scolyer RA. Lymphatic biomarkers in primary melanomas as predictors of regional lymph node metastasis and patient outcomes. Pigment Cell Melanoma Res 2013; 26:326-37. [PMID: 23298266 DOI: 10.1111/pcmr.12064] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 01/02/2013] [Indexed: 11/26/2022]
Abstract
Recently developed lymphatic-specific immunohistochemical markers can now be utilized to assess intratumoral and/or peritumoral lymphatic vessel density (LVD), to detect lymphatic vessel invasion (LVI) by melanoma cells and to identify lymphatic marker expression in melanoma cells themselves. We systematically reviewed the available evidence for the expression of lymphatic markers as predictors of regional node metastasis and survival in melanoma patients. The currently available evidence suggests that LVD (particularly in a peritumoral location) and LVI are predictors of sentinel node metastasis and poorer survival. Nevertheless, adherence to international guidelines in the conduct and reporting of the studies was generally poor, with wide methodologic variations and heterogeneous findings. Larger, carefully conducted and well-reported studies that confirm these preliminary findings are required before it would be appropriate to recommend the routine application of costly and time-consuming immunohistochemistry for lymphatic markers in the routine clinical assessment of primary cutaneous melanomas.
Collapse
Affiliation(s)
- Sandro Pasquali
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.
| | | | | | | | | | | |
Collapse
|
138
|
Phenotype-based high-content chemical library screening identifies statins as inhibitors of in vivo lymphangiogenesis. Proc Natl Acad Sci U S A 2012; 109:E2665-74. [PMID: 22949700 DOI: 10.1073/pnas.1206036109] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lymphangiogenesis plays an important role in promoting cancer metastasis to sentinel lymph nodes and beyond and also promotes organ transplant rejection. We used human lymphatic endothelial cells to establish a reliable three-dimensional lymphangiogenic sprouting assay with automated image acquisition and analysis for inhibitor screening. This high-content phenotype-based assay quantifies sprouts by automated fluorescence microscopy and newly developed analysis software. We identified signaling pathways involved in lymphangiogenic sprouting by screening the Library of Pharmacologically Active Compounds (LOPAC)(1280) collection of pharmacologically relevant compounds. Hit characterization revealed that mitogen-activated protein kinase kinase (MEK) 1/2 inhibitors substantially block lymphangiogenesis in vitro and in vivo. Importantly, the drug class of statins, for the first time, emerged as potent inhibitors of lymphangiogenic sprouting in vitro and of corneal and cutaneous lymphangiogenesis in vivo. This effect was mediated by inhibition of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase and subsequently the isoprenylation of Rac1. Supplementation with the enzymatic products of HMG-CoA reductase functionally rescued lymphangiogenic sprouting and the recruitment of Rac1 to the plasma membrane.
Collapse
|
139
|
Blei F. Update September 2012. Lymphat Res Biol 2012. [DOI: 10.1089/lrb.2012.1035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Francine Blei
- Hassenfeld Children's Center for Cancer and Blood Disorders of NYU Medical Center, New York, New York
| |
Collapse
|
140
|
Alitalo A, Detmar M. Interaction of tumor cells and lymphatic vessels in cancer progression. Oncogene 2011; 31:4499-508. [PMID: 22179834 DOI: 10.1038/onc.2011.602] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastatic spread of cancer through the lymphatic system affects hundreds of thousands of patients yearly. Growth of new lymphatic vessels, lymphangiogenesis, is activated in cancer and inflammation, but is largely inactive in normal physiology, and therefore offers therapeutic potential. Key mediators of lymphangiogenesis have been identified in developmental studies. During embryonic development, lymphatic endothelial cells derive from the blood vascular endothelium and differentiate under the guidance of lymphatic-specific regulators, such as the prospero homeobox 1 transcription factor. Vascular endothelial growth factor-C (VEGF-C) and VEGF receptor 3 signaling are essential for the further development of lymphatic vessels and therefore they provide a promising target for inhibition of tumor lymphangiogenesis. Lymphangiogenesis is important for the progression of solid tumors as shown for melanoma and breast cancer. Tumor cells may use chemokine gradients as guidance cues and enter lymphatic vessels through intercellular openings between endothelial cell junctions or, possibly, by inducing larger discontinuities in the endothelial cell layer. Tumor-draining sentinel lymph nodes show enhanced lymphangiogenesis even before cancer metastasis and they may function as a permissive 'lymphovascular niche' for the survival of metastatic cells. Although our current knowledge indicates that the development of anti-lymphangiogenic therapies may be beneficial for the treatment of cancer patients, several open questions remain with regard to the frequency, mechanisms and biological importance of lymphatic metastases.
Collapse
Affiliation(s)
- A Alitalo
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|