Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-24T15:17:55.112Z Has data issue: false hasContentIssue false

Antiangiogenic and anticancer molecules in cartilage

Published online by Cambridge University Press:  23 April 2012

Debabrata Patra
Affiliation:
Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
Linda J. Sandell*
Affiliation:
Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
*
*Corresponding author: Linda J. Sandell. E-mail: sandelll@wudosis.wustl.edu

Abstract

Cartilage is one of the very few naturally occurring avascular tissues where lack of angiogenesis is the guiding principle for its structure and function. This has attracted investigators who have sought to understand the biochemical basis for its avascular nature, hypothesising that it could be used in designing therapies for treating cancer and related malignancies in humans through antiangiogenic applications. Cartilage encompasses primarily a specialised extracellular matrix synthesised by chondrocytes that is both complex and unique as a result of the myriad molecules of which it is composed. Of these components, a few such as thrombospondin-1, chondromodulin-1, the type XVIII-derived endostatin, SPARC (secreted protein acidic and rich in cysteine) and the type II collagen-derived N-terminal propeptide (PIIBNP) have demonstrated antiangiogenic or antitumour properties in vitro and in vivo preclinical trials that involve several complicated mechanisms that are not completely understood. Thrombospondin-1, endostatin and the shark-cartilage-derived Neovastat preparation have also been investigated in human clinical trials to treat several different kinds of cancers, where, despite the tremendous success seen in preclinical trials, these molecules are yet to show success as anticancer agents. This review summarises the current state-of-the-art antiangiogenic characterisation of these molecules, highlights their most promising aspects and evaluates the future of these molecules in antiangiogenic applications.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Folkman, J. (1971) Tumor angiogenesis: therapeutic implications. New England Journal of Medicine 285, 1182-1186Google Scholar
2Sandell, L.J., Heinegard, D. and Hering, T.M. (2007) Cell biology, biochemistry, and molecular biology of articular cartilage in osteoarthritis. In Osteoarthritis: Diagnosis and Medical/Surgical Management (4th edn), Lippincott Williams & Wilkins, PhiladelphiaGoogle Scholar
3Kronenberg, H.M. (2003) Developmental regulation of the growth plate. Nature 423, 332-336Google Scholar
4Ortega, N., Behonick, D.J. and Werb, Z. (2004) Matrix remodeling during endochondral ossification. Trends in Cell Biology 14, 86-93CrossRefGoogle ScholarPubMed
5Brem, H. and Folkman, J. (1975) Inhibition of tumor angiogenesis mediated by cartilage. Journal of Experimental Medicine 141, 427-439Google Scholar
6Langer, R. et al. (1976) Isolations of a cartilage factor that inhibits tumor neovascularization. Science 193, 70-72Google Scholar
7Cheung, W.H. et al. (2001) Growth plate chondrocytes inhibit neo-angiogenesis – a possible mechanism for tumor control. Cancer Letters 163, 25-32Google Scholar
8Ostrander, G.K. et al. (2004) Shark cartilage, cancer and the growing threat of pseudoscience. Cancer Research 64, 8485-8491Google Scholar
9Dupont E., B.P. and Juneau, C. (1997) Extracts of shark cartilage having an antiangiogenic activity and an effect on tumor progression: process of making thereof. US Patent 5,618,925Google Scholar
10Dupont, E. et al. (2002) Antiangiogenic and antimetastatic properties of Neovastat (AE-941), an orally active extract derived from cartilage tissue. Clinical and Experimental Metastasis 19, 145-153Google Scholar
11Boivin, D. et al. (2002) The antiangiogenic agent Neovastat (AE-941) induces endothelial cell apoptosis. Molecular Cancer Therapeutics 1, 795-802Google Scholar
12Olsson, A.K. et al. (2006) VEGF receptor signalling – in control of vascular function. Nature Reviews Molecular Cell Biology 7, 359-371CrossRefGoogle ScholarPubMed
13Beliveau, R. et al. (2002) The antiangiogenic agent neovastat (AE-941) inhibits vascular endothelial growth factor-mediated biological effects. Clinical Cancer Research 8, 1242-1250Google ScholarPubMed
14Gingras, D. et al. (2001) Matrix proteinase inhibition by AE-941, a multifunctional antiangiogenic compound. Anticancer Research 21, 145-155Google ScholarPubMed
15Weber, M.H., Lee, J. and Orr, F.W. (2002) The effect of Neovastat (AE-941) on an experimental metastatic bone tumor model. International Journal of Oncology 20, 299-303Google Scholar
16Lee, S.Y., Paik, S.Y. and Chung, S.M. (2005) Neovastat (AE-941) inhibits the airway inflammation and hyperresponsiveness in a murine model of asthma. Journal of Microbiology 43, 11-16Google Scholar
17Lee, S.Y. and Chung, S.M. (2007) Neovastat (AE-941) inhibits the airway inflammation via VEGF and HIF-2 alpha suppression. Vascular Pharmacology 47, 313-318Google Scholar
18Nisato, R.E. et al. (2005) Dissecting the role of matrix metalloproteinases (MMP) and integrin alpha(v)beta3 in angiogenesis in vitro: absence of hemopexin C domain bioactivity, but membrane-type 1-MMP and alpha(v)beta3 are critical. Cancer Research 65, 9377-9387Google Scholar
19Pepper, M.S. (2001) Role of the matrix metalloproteinase and plasminogen activator-plasmin systems in angiogenesis. Arteriosclerois, Thrombosis, and Vascular Biology 21, 1104-1117CrossRefGoogle ScholarPubMed
20Lijnen, H.R. and Collen, D. (1995) Mechanisms of physiological fibrinolysis. Baillieres Clinical Haematology 8, 277-290Google Scholar
21Tressler, R.J. et al. (1999) Urokinase receptor antagonists: discovery and application to in vivo models of tumor growth. Acta Pathologica, Microbiologica, et Immunologica Scandinavica 107, 168-173Google Scholar
22Min, H.Y. et al. (1996) Urokinase receptor antagonists inhibit angiogenesis and primary tumor growth in syngeneic mice. Cancer Research 56, 2428-2433Google ScholarPubMed
23Gingras, D. et al. (2004) The antiangiogenic agent Neovastat (AE-941) stimulates tissue plasminogen activator activity. Investigational New Drugs 22, 17-26Google Scholar
24Gingras, D. et al. (2004) Activation of tissue plasminogen activator gene transcription by Neovastat, a multifunctional antiangiogenic agent. Biochemical and Biophysical Research Communications 320, 205-212CrossRefGoogle ScholarPubMed
25Simard, B. et al. (2011) Induction of the fibrinolytic system by cartilage extract mediates its antiangiogenic effect in mouse glioma. Microvascular Research 82, 6-17CrossRefGoogle ScholarPubMed
26Batist, G. et al. (2002) Neovastat (AE-941) in refractory renal cell carcinoma patients: report of a phase II trial with two dose levels. Annals of Oncology 13, 1259-1263Google Scholar
27Latreille, J. et al. (2003) Phase I/II trial of the safety and efficacy of AE-941 (Neovastat) in the treatment of non-small-cell lung cancer. Clinical Lung Cancer 4, 231-236Google Scholar
28Lu, C. et al. (2010) Chemoradiotherapy with or without AE-941 in stage III non-small cell lung cancer: a randomized phase III trial. Journal of the National Cancer Institute 102, 859-865Google Scholar
29White, J. (2010) The challenge of rational development of complex natural products as cancer therapeutics. Journal of the National Cancer Institute 102, 834-835Google Scholar
30Hiraki, Y. et al. (1996) A novel growth-promoting factor derived from fetal bovine cartilage, chondromodulin II. Purification and amino acid sequence. Journal of Biological Chemistry 271, 22657-22662Google Scholar
31Hiraki, Y. et al. (1997) Identification of chondromodulin I as a novel endothelial cell growth inhibitor. Purification and its localization in the avascular zone of epiphyseal cartilage. Journal of Biological Chemistry 272, 32419-32426CrossRefGoogle ScholarPubMed
32Shukunami, C. et al. (1999) Spatiotemporal pattern of the mouse chondromodulin-I gene expression and its regulatory role in vascular invasion into cartilage during endochondral bone formation. International Journal of Developmental Biology 43, 39-49Google Scholar
33Brandau, O. et al. (2001) A novel gene, tendin, is strongly expressed in tendons and ligaments and shows high homology with chondromodulin-I. Developmental Dynamics 221, 72-80Google Scholar
34Yoshioka, M. et al. (2006) Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis. Nature Medicine 12, 1151-1159Google Scholar
35Kusafuka, K. et al. (2002) Cartilage-specific matrix protein, chondromodulin-I (ChM-I), is a strong angio-inhibitor in endochondral ossification of human neonatal vertebral tissues in vivo: relationship with angiogenic factors in the cartilage. Acta Histochemica 104, 167-175Google Scholar
36Brandau, O. et al. (2002) Chondromodulin I is dispensable during enchondral ossification and eye development. Molecular and Cellular Biology 22, 6627-6635Google Scholar
37Funaki, H. et al. (2001) Expression and localization of angiogenic inhibitory factor, chondromodulin-I, in adult rat eye. Investigative Ophthalmology and Visual Science 42, 1193-1200Google ScholarPubMed
38Oshima, Y. et al. (2003) Expression and localization of tenomodulin, a transmembrane type chondromodulin-I-related angiogenesis inhibitor, in mouse eyes. Investigative Ophthalmology and Visual Science 44, 1814-1823Google Scholar
39Docheva, D. et al. (2005) Tenomodulin is necessary for tenocyte proliferation and tendon maturation. Molecular and Cellular Biology 25, 699-705CrossRefGoogle ScholarPubMed
40Ducy, P. et al. (1997) Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89, 747-754Google Scholar
41Komori, T. et al. (1997) Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89, 755-764CrossRefGoogle Scholar
42Otto, F. et al. (1997) Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89, 765-771Google Scholar
43Takeda, S. et al. (2001) Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes and Development 15, 467-481Google Scholar
44Hayami, T. et al. (2003) Expression of the cartilage derived anti-angiogenic factor chondromodulin-I decreases in the early stage of experimental osteoarthritis. Journal of Rheumatology 30, 2207-2217Google ScholarPubMed
45Nagai, T. et al. (2010) Intravenous administration of anti-vascular endothelial growth factor humanized monoclonal antibody bevacizumab improves articular cartilage repair. Arthritis Research and Therapy 12, R178Google Scholar
46Yukata, K. et al. (2008) Altered fracture callus formation in chondromodulin-I deficient mice. Bone 43, 1047-1056Google Scholar
47Blanke, M. et al. (2009) Transplanted chondrocytes inhibit endochondral ossification within cartilage repair tissue. Calcified Tissue International 85, 421-433Google Scholar
48Klinger, P. et al. (2011) Chondromodulin-I stabilizes the chondrocyte phenotype and inhibits endochondral ossification of porcine cartilage repair tissue. Arthritis and Rheumatism 63, 2721-2731CrossRefGoogle Scholar
49Sakamoto, J. et al. (2009) Immobilization-induced cartilage degeneration mediated through expression of hypoxia-inducible factor-1alpha, vascular endothelial growth factor, and chondromodulin-I. Connective Tissue Research 50, 37-45Google Scholar
50Moreno, P.R. et al. (2011) Increased macrophage infiltration and neovascularization in congenital bicuspid aortic valve stenosis. Journal of Thoracic and Cardiovascular Surgery 142, 895-901Google Scholar
51Hayami, T. et al. (1999) Specific loss of chondromodulin-I gene expression in chondrosarcoma and the suppression of tumor angiogenesis and growth by its recombinant protein in vivo. FEBS Letters 458, 436-440Google Scholar
52Mera, H. et al. (2009) Chondromodulin-1 directly suppresses growth of human cancer cells. BMC Cancer 9, 166CrossRefGoogle ScholarPubMed
53Miura, S. et al. (2010) Impairment of VEGF-A-stimulated lamellipodial extensions and motility of vascular endothelial cells by chondromodulin-I, a cartilage-derived angiogenesis inhibitor. Experimental Cell Research 316, 775-788Google Scholar
54Setoguchi, K. et al. (2004) Suppression of T cell responses by chondromodulin I, a cartilage-derived angiogenesis inhibitory factor: therapeutic potential in rheumatoid arthritis. Arthritis and Rheumatism 50, 828-839Google Scholar
55Kyriakides, T.R. et al. (1998) The distribution of the matricellular protein thrombospondin 2 in tissues of embryonic and adult mice. Journal of Histochemistry and Cytochemistry 46, 1007-1015Google Scholar
56Iruela-Arispe, M.L. et al. (1993) Differential expression of thrombospondin 1, 2, and 3 during murine development. Developmental Dynamics 197, 40-56Google Scholar
57Posey, K.L. et al. (2008) Skeletal abnormalities in mice lacking extracellular matrix proteins, thrombospondin-1, thrombospondin-3, thrombospondin-5, and type IX collagen. American Journal of Pathology 172, 1664-1674Google Scholar
58Chen, H., Herndon, M.E. and Lawler, J. (2000) The cell biology of thrombospondin-1. Matrix Biology 19, 597-614Google Scholar
59Lawler, J. (2000) The functions of thrombospondin-1 and-2. Current Opinion in Cell Biology 12, 634-640Google Scholar
60Bornstein, P. (2009) Thrombospondins function as regulators of angiogenesis. Journal of Cell Communication and Signaling 3, 189-200Google Scholar
61Jimenez, B. et al. (2000) Signals leading to apoptosis-dependent inhibition of neovascularization by thrombospondin-1. Nature Medicine 6, 41-48Google Scholar
62Volpert, O.V. et al. (2002) Inducer-stimulated Fas targets activated endothelium for destruction by anti-angiogenic thrombospondin-1 and pigment epithelium-derived factor. Nature Medicine 8, 349-357CrossRefGoogle ScholarPubMed
63Rege, T.A. et al. (2009) Thrombospondin-1-induced apoptosis of brain microvascular endothelial cells can be mediated by TNF-R1. Journal of Cellular Physiology 218, 94-103Google Scholar
64Cursiefen, C. et al. (2011) Thrombospondin 1 inhibits inflammatory lymphangiogenesis by CD36 ligation on monocytes. Journal of Experimental Medicine 208, 1083-1092CrossRefGoogle ScholarPubMed
65Cao, Y. (2005) Opinion: emerging mechanisms of tumour lymphangiogenesis and lymphatic metastasis. Nature Reviews. Cancer 5, 735-743CrossRefGoogle ScholarPubMed
66Gao, P. et al. (2009) Lymphangiogenesis in gastric carcinoma correlates with prognosis. Journal of Pathology 218, 192-200Google Scholar
67Simantov, R., Febbraio, M. and Silverstein, R.L. (2005) The antiangiogenic effect of thrombospondin-2 is mediated by CD36 and modulated by histidine-rich glycoprotein. Matrix Biology 24, 27-34Google Scholar
68Mirochnik, Y., Kwiatek, A. and Volpert, O.V. (2008) Thrombospondin and apoptosis: molecular mechanisms and use for design of complementation treatments. Current Drug Targets 9, 851-862CrossRefGoogle ScholarPubMed
69Yamauchi, M., Imajoh-Ohmi, S. and Shibuya, M. (2007) Novel antiangiogenic pathway of thrombospondin-1 mediated by suppression of the cell cycle. Cancer Science 98, 1491-1497CrossRefGoogle ScholarPubMed
70Oganesian, A. et al. (2008) Thrombospondins use the VLDL receptor and a nonapoptotic pathway to inhibit cell division in microvascular endothelial cells. Molecular Biology of the Cell 19, 563-571Google Scholar
71Chen, H. et al. (1996) Binding and degradation of thrombospondin-1 mediated through heparan sulphate proteoglycans and low-density-lipoprotein receptor-related protein: localization of the functional activity to the trimeric N-terminal heparin-binding region of thrombospondin-1. Biochemical Journal 318, 959-963Google Scholar
72Kawata, K. et al. (2010) Role of the low-density lipoprotein receptor-related protein-1 in regulation of chondrocyte differentiation. Journal of Cellular Physiology 222, 138-148Google Scholar
73Greenaway, J. et al. (2007) Thrombospondin-1 inhibits VEGF levels in the ovary directly by binding and internalization via the low density lipoprotein receptor-related protein-1 (LRP-1). Journal of Cellular Physiology 210, 807-818Google Scholar
74Yang, Z., Strickland, D.K. and Bornstein, P. (2001) Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2. Journal of Biological Chemistry 276, 8403-8408CrossRefGoogle ScholarPubMed
75Hahn-Dantona, E. et al. (2001) The low density lipoprotein receptor-related protein modulates levels of matrix metalloproteinase 9 (MMP-9) by mediating its cellular catabolism. Journal of Biological Chemistry 276, 15498-15503CrossRefGoogle ScholarPubMed
76Hankenson, K.D. et al. (2000) Increased marrow-derived osteoprogenitor cells and endosteal bone formation in mice lacking thrombospondin 2. Journal of Bone and Mineral Research 15, 851-862Google Scholar
77Wang, S. et al. (2003) Thrombospondin-1-deficient mice exhibit increased vascular density during retinal vascular development and are less sensitive to hyperoxia-mediated vessel obliteration. Developmental Dynamics 228, 630-642Google Scholar
78Malek, M.H. and Olfert, I.M. (2009) Global deletion of thrombospondin-1 increases cardiac and skeletal muscle capillarity and exercise capacity in mice. Experimental Physiology 94, 749-760Google Scholar
79Jou, I.M. et al. (2005) Thrombospondin 1 as an effective gene therapeutic strategy in collagen-induced arthritis. Arthritis and Rheumatism 52, 339-344Google Scholar
80Taylor, D.K. et al. (2009) Thrombospondin-2 influences the proportion of cartilage and bone during fracture healing. Journal of Bone Mineral Research 24, 1043-1054Google Scholar
81Gelse, K. et al. (2011) Thrombospondin-1 prevents excessive ossification in cartilage repair tissue induced by osteogenic protein-1. Tissue Engineering Part A 17, 2101-2112Google Scholar
82Hsieh, J.L. et al. (2010) Intraarticular gene transfer of thrombospondin-1 suppresses the disease progression of experimental osteoarthritis. Journal of Orthopaedic Research 28, 1300-1306CrossRefGoogle ScholarPubMed
83Guo, X. et al. (2006) Repair of full-thickness articular cartilage defects by cultured mesenchymal stem cells transfected with the transforming growth factor beta1 gene. Biomedical Materials 1, 206-215CrossRefGoogle ScholarPubMed
84Yamane, S. and Reddi, A.H. (2008) Induction of chondrogenesis and superficial zone protein accumulation in synovial side population cells by BMP-7 and TGF-beta1. Journal of Orthopaedic Research 26, 485-492Google Scholar
85Carlson, C.B., Lawler, J. and Mosher, D.F. (2008) Structures of thrombospondins. Cellular and Molecular Life Sciences 65, 672-686Google Scholar
86Zhang, X. and Lawler, J. (2007) Thrombospondin-based antiangiogenic therapy. Microvascular Research 74, 90-99Google Scholar
87Ren, B. et al. (2006) Regulation of tumor angiogenesis by thrombospondin-1. Biochimica et Biophysica Acta 1765, 178-188Google Scholar
88Kazerounian, S., Yee, K.O. and Lawler, J. (2008) Thrombospondins in cancer. Cellular and Molecular Life Sciences 65, 700-712Google Scholar
89Olsson, A. et al. (2010) Tasquinimod (ABR-215050), a quinoline-3-carboxamide anti-angiogenic agent, modulates the expression of thrombospondin-1 in human prostate tumors. Molecular Cancer 9, 107Google Scholar
90Stapleton, C.J. et al. (2011) Thrombospondin-1 modulates the angiogenic phenotype of human cerebral arteriovenous malformation endothelial cells. Neurosurgery 68, 1342-1353Google Scholar
91Morgan-Rowe, L. et al. (2011) Thrombospondin 1 in hypoxia-conditioned media blocks the growth of human microvascular endothelial cells and is increased in systemic sclerosis tissues. Fibrogenesis Tissue Repair 4, 13CrossRefGoogle ScholarPubMed
92Koch, M. et al. (2011) CD36-mediated activation of endothelial cell apoptosis by an N-terminal recombinant fragment of thrombospondin-2 inhibits breast cancer growth and metastasis in vivo. Breast Cancer Research and Treatment 128, 337-346Google Scholar
93Reiher, F.K. et al. (2002) Inhibition of tumor growth by systemic treatment with thrombospondin-1 peptide mimetics. International Journal of Cancer 98, 682-689CrossRefGoogle ScholarPubMed
94Haviv, F. et al. (2005) Thrombospondin-1 mimetic peptide inhibitors of angiogenesis and tumor growth: design, synthesis, and optimization of pharmacokinetics and biological activities. Journal of Medicinal Chemistry 48, 2838-2846Google Scholar
95Rusk, A. et al. (2006) Preclinical evaluation of antiangiogenic thrombospondin-1 peptide mimetics, ABT-526 and ABT-510, in companion dogs with naturally occurring cancers. Clinical Cancer Research 12, 7444-7455CrossRefGoogle ScholarPubMed
96Rusk, A. et al. (2006) Cooperative activity of cytotoxic chemotherapy with antiangiogenic thrombospondin-I peptides, ABT-526 in pet dogs with relapsed lymphoma. Clinical Cancer Research 12, 7456-7464Google Scholar
97Anderson, J.C. et al. (2007) ABT-510, a modified type 1 repeat peptide of thrombospondin, inhibits malignant glioma growth in vivo by inhibiting angiogenesis. Cancer Biology and Therapy 6, 454-462Google Scholar
98Yang, Q. et al. (2007) Thrombospondin-1 peptide ABT-510 combined with valproic acid is an effective antiangiogenesis strategy in neuroblastoma. Cancer Research 67, 1716-1724Google Scholar
99Campbell, N.E. et al. (2010) The thrombospondin-1 mimetic ABT-510 increases the uptake and effectiveness of cisplatin and paclitaxel in a mouse model of epithelial ovarian cancer. Neoplasia 12, 275-283CrossRefGoogle Scholar
100Hasina, R. et al. (2009) ABT-510 is an effective chemopreventive agent in the mouse 4-nitroquinoline 1-oxide model of oral carcinogenesis. Cancer Prevention Research (Philadelphia) 2, 385-393Google Scholar
101Greenaway, J. et al. (2009) ABT-510 induces tumor cell apoptosis and inhibits ovarian tumor growth in an orthotopic, syngeneic model of epithelial ovarian cancer. Molecular Cancer Therapeutics 8, 64-74Google Scholar
102Hoekstra, R. et al. (2005) Phase I safety, pharmacokinetic, and pharmacodynamic study of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 in patients with advanced cancer. Journal of Clinical Oncology 23, 5188-5197Google Scholar
103Gordon, M.S. et al. (2008) A phase 1 trial of 2 dose schedules of ABT-510, an antiangiogenic, thrombospondin-1-mimetic peptide, in patients with advanced cancer. Cancer 113, 3420-3429CrossRefGoogle ScholarPubMed
104Markovic, S.N. et al. (2007) A phase II study of ABT-510 (thrombospondin-1 analog) for the treatment of metastatic melanoma. American Journal of Clinical Oncology 30, 303-309Google Scholar
105Ebbinghaus, S. et al. (2007) Phase 2 study of ABT-510 in patients with previously untreated advanced renal cell carcinoma. Clinical Cancer Research 13, 6689-6695Google Scholar
106Baker, L.H. et al. (2008) Randomized, phase II study of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 in patients with advanced soft tissue sarcoma. Journal of Clinical Oncology 26, 5583-5588Google Scholar
107Gietema, J.A. et al. (2006) A phase I study assessing the safety and pharmacokinetics of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 with gemcitabine and cisplatin in patients with solid tumors. Annals of Oncology 17, 1320-1327CrossRefGoogle ScholarPubMed
108Hoekstra, R. et al. (2006) Phase I study of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 with 5-fluorouracil and leucovorin: a safe combination. European Journal of Cancer 42, 467-472CrossRefGoogle ScholarPubMed
109Nabors, L.B. et al. (2010) A phase 1 trial of ABT-510 concurrent with standard chemoradiation for patients with newly diagnosed glioblastoma. Archives of Neurology 67, 313-319Google Scholar
110Ferreras, M. et al. (2000) Generation and degradation of human endostatin proteins by various proteinases. FEBS Letters 486, 247-251CrossRefGoogle ScholarPubMed
111Nilsson, U.W. and Dabrosin, C. (2006) Estradiol and tamoxifen regulate endostatin generation via matrix metalloproteinase activity in breast cancer in vivo. Cancer Research 66, 4789-4794Google Scholar
112Pufe, T. et al. (2004) Endostatin/collagen XVIII – an inhibitor of angiogenesis – is expressed in cartilage and fibrocartilage. Matrix Biology 23, 267-276Google Scholar
113Stempel, J. et al. (2011) Development of articular cartilage and the metaphyseal growth plate: the localization of TRAP cells, VEGF, and endostatin. Journal of Anatomy 218, 608-618Google Scholar
114Tan, H. et al. (2011) Down-regulation of vascular endothelial growth factor and up-regulation of pigment epithelium derived factor make low molecular weight heparin-endostatin and polyethylene glycol-endostatin potential candidates for anti-angiogenesis drug. Biological and Pharmaceutical Bulletin 34, 545-550Google Scholar
115Xu, W. et al. (2010) Endostar, a recently introduced recombinant human endostatin, inhibits proliferation and migration through regulating growth factors, adhesion factors and inflammatory mediators in choroid-retinal endothelial cells. Molekuliarnaia Biologiia 44, 664-670Google Scholar
116Yang, L. et al. (2011) Antitumor effect of endostatin overexpressed in C6 glioma cells is associated with the down-regulation of VEGF. International Journal of Oncology 38, 465-471Google Scholar
117Li, Z.Y. et al. (2011) Sp1 inhibition-mediated upregulation of VEGF(165)b induced by rh-endostatin enhances antiangiogenic and anticancer effect of rh-endostatin in A549. Tumour Biology 32, 677-687Google Scholar
118Kim, Y.M. et al. (2002) Endostatin blocks vascular endothelial growth factor-mediated signaling via direct interaction with KDR/Flk-1. Journal of Biological Chemistry 277, 27872-27879Google Scholar
119Karumanchi, S.A. et al. (2001) Cell surface glypicans are low-affinity endostatin receptors. Molecular Cell 7, 811-822CrossRefGoogle ScholarPubMed
120Shi, H. et al. (2007) Nucleolin is a receptor that mediates antiangiogenic and antitumor activity of endostatin. Blood 110, 2899-2906Google Scholar
121Fu, Y. et al. (2009) The heparin binding motif of endostatin mediates its interaction with receptor nucleolin. Biochemistry 48, 11655-11663CrossRefGoogle ScholarPubMed
122Zhuo, W. et al. (2010) Endostatin inhibits tumour lymphangiogenesis and lymphatic metastasis via cell surface nucleolin on lymphangiogenic endothelial cells. Journal of Pathology 222, 249-260CrossRefGoogle ScholarPubMed
123Wickstrom, S.A., Alitalo, K. and Keski-Oja, J. (2003) Endostatin associates with lipid rafts and induces reorganization of the actin cytoskeleton via down-regulation of RhoA activity. Journal of Biological Chemistry 278, 37895-37901Google Scholar
124Wickstrom, S.A., Alitalo, K. and Keski-Oja, J. (2002) Endostatin associates with integrin alpha5beta1 and caveolin-1, and activates Src via a tyrosyl phosphatase-dependent pathway in human endothelial cells. Cancer Research 62, 5580-5589Google Scholar
125Kim, Y.M. et al. (2000) Endostatin inhibits endothelial and tumor cellular invasion by blocking the activation and catalytic activity of matrix metalloproteinase. Cancer Research 60, 5410-5413Google Scholar
126Lee, S.J. et al. (2002) Endostatin binds to the catalytic domain of matrix metalloproteinase-2. FEBS Letters 519, 147-152Google Scholar
127Wickstrom, S.A. et al. (2001) Endostatin-induced modulation of plasminogen activation with concomitant loss of focal adhesions and actin stress fibers in cultured human endothelial cells. Cancer Research 61, 6511-6516Google Scholar
128Dhanabal, M. et al. (1999) Endostatin induces endothelial cell apoptosis. Journal of Biological Chemistry 274, 11721-11726CrossRefGoogle ScholarPubMed
129Hanai, J. et al. (2002) Endostatin is a potential inhibitor of Wnt signaling. Journal of Cell Biology 158, 529-539Google Scholar
130Hanai, J. et al. (2002) Endostatin causes G1 arrest of endothelial cells through inhibition of cyclin D1. Journal of Biological Chemistry 277, 16464-16469Google Scholar
131Nguyen, T.M. et al. (2009) Endostatin induces autophagy in endothelial cells by modulating Beclin 1 and beta-catenin levels. Journal of Cellular and Molecular Medicine 13, 3687-3698Google Scholar
132Dixelius, J. et al. (2000) Endostatin-induced tyrosine kinase signaling through the Shb adaptor protein regulates endothelial cell apoptosis. Blood 95, 3403-3411Google Scholar
133Tolstanova, G. et al. (2011) Role of anti-angiogenic factor endostatin in the pathogenesis of experimental ulcerative colitis. Life Sciences 88, 74-81Google Scholar
134Belotti, D. et al. (2008) Vascular endothelial growth factor stimulates organ-specific host matrix metalloproteinase-9 expression and ovarian cancer invasion. Molecular Cancer Research 6, 525-534CrossRefGoogle ScholarPubMed
135Abdollahi, A. et al. (2004) Endostatin's antiangiogenic signaling network. Molecular Cell 13, 649-663Google Scholar
136Yokoyama, Y. and Ramakrishnan, S. (2004) Improved biological activity of a mutant endostatin containing a single amino-acid substitution. British Journal of Cancer 90, 1627-1635Google Scholar
137Ding, Y.H. et al. (1998) Zinc-dependent dimers observed in crystals of human endostatin. Proceedings of the National Academy of Sciences of the United States of America 95, 10443-10448Google Scholar
138Hohenester, E. et al. (2000) Variable zinc coordination in endostatin. Journal of Molecular Biology 297, 1-6Google Scholar
139Han, Q. et al. (2007) Contributions of Zn(II)-binding to the structural stability of endostatin. FEBS Letters 581, 3027-3032Google Scholar
140Fu, Y. and Luo, Y. (2010) The N-terminal integrity is critical for the stability and biological functions of endostatin. Biochemistry 49, 6420-6429CrossRefGoogle ScholarPubMed
141Yokoyama, Y. and Ramakrishnan, S. (2004) Addition of integrin binding sequence to a mutant human endostatin improves inhibition of tumor growth. International Journal of Cancer 111, 839-848CrossRefGoogle ScholarPubMed
142Jing, Y. et al. (2011) Inhibition of ovarian cancer by RGD-P125A-endostatin-Fc fusion proteins. International Journal of Cancer 129, 751-761Google Scholar
143Chen, C.T. et al. (2011) Dual targeting of tumor angiogenesis and chemotherapy by endostatin-cytosine deaminase-uracil phosphoribosyl transferase. Molecular Cancer Therapeutics 10, 1327-1336Google Scholar
144Shin, S.U. et al. (2011) Targeted delivery of an antibody-mutant human endostatin fusion protein results in enhanced antitumor efficacy. Molecular Cancer Therapeutics 10, 603-614CrossRefGoogle ScholarPubMed
145Paek, S.Y., Kim, Y.S. and Choi, S.G. (2010) The orientation-dependent expression of angiostatin-endostatin hybrid proteins and their characterization for the synergistic effects of antiangiogenesis. Journal of Microbiology and Biotechnology 20, 1430-1435Google Scholar
146Zhao, P. et al. (2008) E10A, an adenovirus carrying human endostatin gene, in combination with docetaxel treatment inhibits prostate cancer growth and metastases. Journal of Cellular and Molecular Medicine 14, 381-391Google Scholar
147Pan, J.G. et al. (2011) Suppression of bladder cancer growth in mice by adeno-associated virus vector-mediated endostatin expression. Tumour Biology 32, 301-310Google Scholar
148Zhu, G. et al. (2011) Glioma stem cells targeted by oncolytic virus carrying endostatin–angiostatin fusion gene and the expression of its exogenous gene in vitro. Brain Research 1390, 59-69Google Scholar
149Jiang, J. et al. (2010) The effect of endostatin mediated by human mesenchymal stem cells on ovarian cancer cells in vitro. Journal of Cancer Research and Clinical Oncology 136, 873-881Google Scholar
150Tysome, J.R. et al. (2011) Lister vaccine strain of vaccinia virus armed with the endostatin–angiostatin fusion gene: an oncolytic virus superior to dl1520 (ONYX-015) for human head and neck cancer. Human Gene Therapy 22, 1101-1108CrossRefGoogle ScholarPubMed
151Yin, J. et al. (2011) hMSC-mediated concurrent delivery of endostatin and carboxylesterase to mouse xenografts suppresses glioma initiation and recurrence. Molecular Therapy 19, 1161-1169Google Scholar
152Coutinho, E.L. et al. (2007) Anti-tumor effect of endostatin mediated by retroviral gene transfer in mice bearing renal cell carcinoma. FASEB Journal 21, 3153-3161Google Scholar
153Zhang, D.W. et al. (2010) The synergistic effect of recombinant human endostatin (YH-16) combined with oxaliplatin on human colorectal carcinoma. Journal of International Medical Research 38, 111-126Google Scholar
154Huang, G. and Chen, L. (2010) Recombinant human endostatin improves anti-tumor efficacy of paclitaxel by normalizing tumor vasculature in Lewis lung carcinoma. Journal of Cancer Research and Clinical Oncology 136, 1201-1211Google Scholar
155Jiang, X.D. et al. (2011) Inhibitory effect of radiotherapy combined with weekly recombinant human endostatin on the human pulmonary adenocarcinoma A549 xenografts in nude mice. Lung Cancer 72, 165-171Google Scholar
156Peng, X.C. et al. (2011) Different combination schedules of gemcitabine with endostar affect antitumor efficacy. Cancer Chemotherapy and Pharmacology 69, 239-246Google Scholar
157Jia, Y. et al. (2011) Recombinant human endostatin, Endostar, enhances the effects of chemo-radiotherapy in a mouse cervical cancer xenograft model. European Journal of Gynaecological Oncology 32, 316-324Google Scholar
158Li, X.Q. et al. (2011) Endostar, a modified recombinant human endostatin, exhibits synergistic effects with dexamethasone on angiogenesis and hepatoma growth. Cancer Letters 301, 212-220Google Scholar
159Ou, J. et al. (2011) Endostatin suppresses colorectal tumor-induced lymphangiogenesis by inhibiting expression of fibronectin extra domain a and integrin alpha9. Journal of Cellular Biochemistry 112, 2106-2114Google Scholar
160Jeng, L., Olsen, B.R. and Spector, M. (2010) Engineering endostatin-producing cartilaginous constructs for cartilage repair using nonviral transfection of chondrocyte-seeded and mesenchymal-stem-cell-seeded collagen scaffolds. Tissue Engineering Part A 16, 3011-3021CrossRefGoogle ScholarPubMed
161Feng, Y. et al. (2005) Endostatin promotes the anabolic program of rabbit chondrocyte. Cell Research 15, 201-206Google Scholar
162Fu, Y. et al. (2009) Unraveling the mysteries of endostatin. IUBMB Life 61, 613-626CrossRefGoogle ScholarPubMed
163Kulke, M.H. et al. (2006) Phase II study of recombinant human endostatin in patients with advanced neuroendocrine tumors. Journal of Clinical Oncology 24, 3555-3561Google Scholar
164Eder, J.P. Jr et al. (2002) Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily. Journal of Clinical Oncology 20, 3772-3784Google Scholar
165Herbst, R.S. et al. (2002) Phase I study of recombinant human endostatin in patients with advanced solid tumors. Journal of Clinical Oncology 20, 3792-3803Google Scholar
166Thomas, J.P. et al. (2003) Phase I pharmacokinetic and pharmacodynamic study of recombinant human endostatin in patients with advanced solid tumors. Journal of Clinical Oncology 21, 223-231Google Scholar
167Hansma, A.H. et al. (2005) Recombinant human endostatin administered as a 28-day continuous intravenous infusion, followed by daily subcutaneous injections: a phase I and pharmacokinetic study in patients with advanced cancer. Annals of Oncology 16, 1695-1701Google Scholar
168Li, Y. et al. (2010) Efficacy and safety of endostar combined with chemotherapy in patients with advanced solid tumors. Asian Pacific Journal of Cancer Prevention 11, 1119-1123Google Scholar
169Zhou, Z.T. et al. (2011) Phase II study of cisplatin/etoposide and endostar for extensive-stage small-cell lung cancer. Cancer Chemotherapy and Pharmacology 68, 1027-1032Google Scholar
170Zhao, X. et al. (2011) A randomized phase II study of recombinant human endostatin plus gemcitabine/cisplatin compared with gemcitabine/cisplatin alone as first-line therapy in advanced non-small-cell lung cancer. Investigational New Drugs (Epub ahead of print, doi:10.1007/s10637-011-9631-7)Google Scholar
171Han, B. et al. (2011) A multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy of paclitaxel-carboplatin alone or with endostar for advanced non-small cell lung cancer. Journal of Thoracic Oncology 6, 1104-1109CrossRefGoogle ScholarPubMed
172Podhajcer, O.L. et al. (2008) The role of the matricellular protein SPARC in the dynamic interaction between the tumor and the host. Cancer Metastasis Reviews 27, 691-705Google Scholar
173Nagaraju, G.P. and Sharma, D. (2011) Anti-cancer role of SPARC, an inhibitor of adipogenesis. Cancer Treat Reviews 37, 559-566Google Scholar
174Arnold, S.A. and Brekken, R.A. (2009) SPARC: a matricellular regulator of tumorigenesis. Journal of Cell Communication and Signaling 3, 255-273Google Scholar
175Termine, J.D. et al. (1981) Osteonectin, a bone-specific protein linking mineral to collagen. Cell 26, 99-105Google Scholar
176Chandrasekhar, S. et al. (1994) Osteonectin/SPARC is a product of articular chondrocytes/cartilage and is regulated by cytokines and growth factors. Biochimica et Biophysica Acta 1221, 7-14Google Scholar
177Copray, J.C. et al. (1989) Presence of osteonectin/SPARC in mandibular condylar cartilage of the rat. Journal of Anatomy 162, 43-51Google Scholar
178Hecht, J.T. and Sage, E.H. (2006) Retention of the matricellular protein SPARC in the endoplasmic reticulum of chondrocytes from patients with pseudoachondroplasia. Journal of Histochemistry Cytochemistry 54, 269-74Google Scholar
179Chlenski, A. et al. (2006) SPARC expression is associated with impaired tumor growth, inhibited angiogenesis and changes in the extracellular matrix. International Journal of Cancer 118, 310-316Google Scholar
180Yiu, G.K. et al. (2001) SPARC (secreted protein acidic and rich in cysteine) induces apoptosis in ovarian cancer cells. American Journal of Pathology 159, 609-622Google Scholar
181Dhanesuan, N. et al. (2002) Doxycycline-inducible expression of SPARC/Osteonectin/BM40 in MDA-MB-231 human breast cancer cells results in growth inhibition. Breast Cancer Research and Treatment 75, 73-85Google Scholar
182Chlenski, A. et al. (2004) Neuroblastoma angiogenesis is inhibited with a folded synthetic molecule corresponding to the epidermal growth factor-like module of the follistatin domain of SPARC. Cancer Research 64, 7420-7425Google Scholar
183Hasselaar, P. and Sage, E.H. (1992) SPARC antagonizes the effect of basic fibroblast growth factor on the migration of bovine aortic endothelial cells. Journal of Cellular Biochemistry 49, 272-283Google Scholar
184Raines, E.W. et al. (1992) The extracellular glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-AB and -BB and inhibits the binding of PDGF to its receptors. Proceedings of the National Academy of Sciences of the United States of America 89, 1281-1285Google Scholar
185Kupprion, C., Motamed, K. and Sage, E.H. (1998) SPARC (BM-40, osteonectin) inhibits the mitogenic effect of vascular endothelial growth factor on microvascular endothelial cells. Journal of Biological Chemistry 273, 29635-29640Google Scholar
186Norose, K. et al. (2000) Lenses of SPARC-null mice exhibit an abnormal cell surface-basement membrane interface. Experimental Eye Research 71, 295-307CrossRefGoogle ScholarPubMed
187Liang, J.F. et al. (2010) Relationship and prognostic significance of SPARC and VEGF protein expression in colon cancer. Journal of Experimental and Clinical Cancer Research 29, 71Google Scholar
188Chew, A. et al. (2011) SPARC, FOXP3, CD8 and CD45 Correlation with Disease Recurrence and Long-Term Disease-Free Survival in Colorectal Cancer. PLoS One 6, e22047Google Scholar
189Wang, C.S. et al. (2004) Overexpression of SPARC gene in human gastric carcinoma and its clinic-pathologic significance. British Journal of Cancer 91, 1924-1930Google Scholar
190Yamashita, K. et al. (2003) Clinical significance of secreted protein acidic and rich in cystein in esophageal carcinoma and its relation to carcinoma progression. Cancer 97, 2412-2419Google Scholar
191Prockop, D.J., Sieron, A.L. and Li, S.W. (1998) Procollagen N-proteinase and procollagen C-proteinase. Two unusual metalloproteinases that are essential for procollagen processing probably have important roles in development and cell signaling. Matrix Biology 16, 399-408Google Scholar
192Sandell, L.J. et al. (1991) Alternatively spliced type II procollagen mRNAs define distinct populations of cells during vertebral development: differential expression of the amino-propeptide. Journal of Cell Biology. 114, 1307-1319Google Scholar
193Zhu, Y. et al. (1999) Type IIA procollagen containing the cysteine-rich amino propeptide is deposited in the extracellular matrix of prechondrogenic tissue and binds to TGF-beta1 and BMP-2. Journal of Cell Biology. 144, 1069-1080Google Scholar
194Wang, Z. et al. (2010) Type IIB procollagen NH(2)-propeptide induces death of tumor cells via interaction with integrins alpha(V)beta(3) and alpha(V)beta(5). Journal of Biological Chemistry 285, 20806-20817Google Scholar
195Hayashi, S. et al. (2011) The type II collagen N-propeptide, PIIBNP, inhibits cell survival and bone resorption of osteoclasts via integrin-mediated signaling. Bone 49, 644-652Google Scholar
196Allegra, C.J. et al. (2011) Phase III trial assessing bevacizumab in stages II and III carcinoma of the colon: results of NSABP protocol C-08. Journal of Clinical Oncology 29, 11-16Google Scholar
197Van Cutsem, E. et al. (2009) Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. Journal of Clinical Oncology 27, 2231-2237Google Scholar
198Casanovas, O. et al. (2005) Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8, 299-309Google Scholar
199Shojaei, F. et al. (2007) Tumor refractoriness to anti-VEGF treatment is mediated by CD11b + Gr1+ myeloid cells. Nature Biotechnology 25, 911-920Google Scholar
200Syrigos, K.N. et al. (2011) Bevacizumab-induced hypertension: pathogenesis and management. BioDrugs 25, 159-169Google Scholar
201Boivin, D. et al. (2004) Purification and characterization of a stimulator of plasmin generation from the antiangiogenic agent Neovastat: identification as immunoglobulin kappa light chain. Archives of Biochemistry and Biophysics 431, 197-206Google Scholar
202Rabbani-Chadegani, A. et al. (2008) Identification of low-molecular-weight protein (SCP1) from shark cartilage with anti-angiogenesis activity and sequence similarity to parvalbumin. Journal of Pharmaceutical and Biomedical Analysis 46, 563-567Google Scholar
203Zheng, L. et al. (2007) A novel polypeptide from shark cartilage with potent anti-angiogenic activity. Cancer Biology and Therapy 6, 775-780Google Scholar
204Bargahi, A. and Rabbani-Chadegani, A. (2008) Angiogenic inhibitor protein fractions derived from shark cartilage. Bioscience Reports 28, 15-21Google Scholar
205Johnson, W.E. et al. (2005) Human intervertebral disc aggrecan inhibits endothelial cell adhesion and cell migration in vitro. Spine 30, 1139-1147Google Scholar
206Liu, A. et al. (2009) Cartilage polysaccharide induces apoptotic cell death of L1210 cells. Leukemia and Lymphoma 50, 1017-1029Google Scholar
207Bara, J.J. et al. (2011) Articular cartilage glycosaminoglycans inhibit the adhesion of endothelial cells. Connective Tissue Research (Epub ahead of print, doi:10.3109/03008207.2011.629310)Google Scholar
208Moses, M.A. et al. (1999) Troponin I is present in human cartilage and inhibits angiogenesis. Proceedings of the National Academy of Sciences of the United States of America 96, 2645-2650Google Scholar
209Feldman, L. and Rouleau, C. (2002) Troponin I inhibits capillary endothelial cell proliferation by interaction with the cell's bFGF receptor. Microvascular Research 63, 41-49Google Scholar
210Liu, N. et al. (2001) Metastatin: a hyaluronan-binding complex from cartilage that inhibits tumor growth. Cancer Research 61, 1022-1028Google Scholar
211Macotela, Y. et al. (2006) Matrix metalloproteases from chondrocytes generate an antiangiogenic 16 kDa prolactin. Journal of Cell Science 119, 1790-1800CrossRefGoogle ScholarPubMed
212Donnini, S. et al. (2007) Pyrazolo-pyrimidine-derived c-Src inhibitor reduces angiogenesis and survival of squamous carcinoma cells by suppressing vascular endothelial growth factor production and signaling. International Journal of Cancer 120, 995-1004Google Scholar
213Tong, Y. et al. (2010) Characterization of a monoPEG20000-Endostar. International Journal of Biological Macromolecules 46, 331-336Google Scholar
214Xu, H.M. et al. (2008) An RGD-modified endostatin-derived synthetic peptide shows antitumor activity in vivo. Bioconjugate Chemistry 19, 1980-1986Google Scholar
215Yin, R. et al. (2010) Effect of RGD-4C position is more important than disulfide bonds on antiangiogenic activity of RGD-4C modified endostatin derived synthetic polypeptide. Bioconjugate Chemistry 21, 1142-1147Google Scholar
216Hai-Tao, Z. et al. (2011) A tumor-penetrating peptide modification enhances the antitumor activity of endostatin in vivo. Anticancer Drugs 22, 409-415Google Scholar
217Wang, L. et al. (2011) Gene therapy with recombinant adenovirus encoding endostatin encapsulated in cationic liposome in coxsackievirus and adenovirus receptor-deficient colon carcinoma murine models. Human Gene Therapy 22, 1061-1069Google Scholar
218Yang, L. et al. (2010) Suppression of ovarian cancer growth via systemic administration with liposome-encapsulated adenovirus-encoding endostatin. Cancer Gene Therapy 17, 49-57Google Scholar

Further reading, resources and contacts

For detailed information on clinical trials using anti-VEGF therapies, endostatin or the TSP1-derived ABT-510 in treatment of cancer, see:

Bonnet, C.S. and Walsh, D.A. (2005) Osteoarthritis, angiogenesis and inflammation. Rheumatology 44, 7-16Google Scholar
Zheng, M.-J. (2009) Endostatin derivative angiogenesis inhibitors. Chinese Medical Journal 122, 1947-1951Google Scholar
Rosca, E.V. et al. (2011) Anti-angiogenic peptides for cancer therapeutics. Current Pharmaceutical Biotechnology 12, 1101-1116Google Scholar
Wang, J. et al. (2005) Results of randomized, multicenter, double-blind phase III trial of recombinant human endostatin (YH-16) in treatment of non-small cell lung cancer patients. Zhongguo Fei Ai Za Zhi 8, 283-290Google Scholar
Faye, C. et al. (2009) The first draft of the endostatin interaction network. Journal of Biological Chemistry 284, 22041-22047Google Scholar
Goel, S. et al. (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiological Reviews 91, 1071-1121Google Scholar
Sato, Y. (2011) Persistent vascular normalization as an alternative goal of anti-angiogenic cancer therapy. Cancer Science 102, 1253-1256Google Scholar
Minchinton, A.I. and Tannock, I.F. (2006) Drug penetration in solid tumours. Nature Reviews Cancer 6, 583-592Google Scholar
Yan, Q. and Sage, E.H. (1999) SPARC, a matricellular glycoprotein with important biological functions. Journal of Histochemistry and Cytochemistry 47, 1495-1505Google Scholar
Bradshaw, A.D. and Sage, E.H. (2001) SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury. Journal of Clinical Investigation 107, 1049-1054Google Scholar
Bradshaw, A.D. (2009) The role of SPARC in extracellular matrix assembly. Journal of Cell Communication and Signaling 3, 239-246Google Scholar
Bonnet, C.S. and Walsh, D.A. (2005) Osteoarthritis, angiogenesis and inflammation. Rheumatology 44, 7-16Google Scholar
Zheng, M.-J. (2009) Endostatin derivative angiogenesis inhibitors. Chinese Medical Journal 122, 1947-1951Google Scholar
Rosca, E.V. et al. (2011) Anti-angiogenic peptides for cancer therapeutics. Current Pharmaceutical Biotechnology 12, 1101-1116Google Scholar
Wang, J. et al. (2005) Results of randomized, multicenter, double-blind phase III trial of recombinant human endostatin (YH-16) in treatment of non-small cell lung cancer patients. Zhongguo Fei Ai Za Zhi 8, 283-290Google Scholar
Faye, C. et al. (2009) The first draft of the endostatin interaction network. Journal of Biological Chemistry 284, 22041-22047Google Scholar
Goel, S. et al. (2011) Normalization of the vasculature for treatment of cancer and other diseases. Physiological Reviews 91, 1071-1121Google Scholar
Sato, Y. (2011) Persistent vascular normalization as an alternative goal of anti-angiogenic cancer therapy. Cancer Science 102, 1253-1256Google Scholar
Minchinton, A.I. and Tannock, I.F. (2006) Drug penetration in solid tumours. Nature Reviews Cancer 6, 583-592Google Scholar
Yan, Q. and Sage, E.H. (1999) SPARC, a matricellular glycoprotein with important biological functions. Journal of Histochemistry and Cytochemistry 47, 1495-1505Google Scholar
Bradshaw, A.D. and Sage, E.H. (2001) SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury. Journal of Clinical Investigation 107, 1049-1054Google Scholar
Bradshaw, A.D. (2009) The role of SPARC in extracellular matrix assembly. Journal of Cell Communication and Signaling 3, 239-246Google Scholar