Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 90(12): 2418-24, 2004 Jun 14.
Article in English | MEDLINE | ID: mdl-15173857

ABSTRACT

The antineoplastic compound aplidine, a new marine-derived depsipeptide, has shown preclinical activity in vitro on haematological and solid tumour cell lines. It is currently in early phase clinical trials. The exact mechanism of action of this anticancer agent still needs to be clarified. We have previously reported that aplidine blocks the secretion of the angiogenic factor vascular endothelial growth factor (VEGF) by the human leukaemia cells MOLT-4, suggesting a possible effect on tumour angiogenesis. This study was designed to investigate the antiangiogenic effect of aplidine. In vivo, in the chick embryo allantoic membrane (CAM) assay, aplidine inhibited spontaneous angiogenesis, angiogenesis elicited by exogenous VEGF and FGF-2, and induced by VEGF overexpressing 1A9 ovarian carcinoma cells. In vitro, at concentrations achievable in the plasma of patients, aplidine inhibited endothelial cell functions related to angiogenesis. It affected VEGF- and FGF-2-induced endothelial cell proliferation, inhibited cell migration and invasiveness assessed in the Boyden chamber and blocked the production of matrix metalloproteinases (MMP-2 and MMP-9) by endothelial cells. Finally, aplidine prevented the formation of capillary-like structures by endothelial cells on Matrigel. These findings indicate that aplidine has antiangiogenic activity in vivo and inhibits endothelial cell functional responses to angiogenic stimuli in vitro. This effect might contribute to the antineoplastic activity of aplidine.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Depsipeptides , Neovascularization, Pathologic , Peptides, Cyclic/pharmacology , Animals , Biological Assay , Capillaries , Cell Culture Techniques , Chick Embryo , Endothelial Cells , Humans , Matrix Metalloproteinases/analysis , Umbilical Veins/cytology
2.
Leukemia ; 17(1): 52-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12529660

ABSTRACT

The mechanism by which aplidine, a marine natural product in early clinical development as an anticancer agent, induces cell growth inhibition and apoptosis has been investigated in the human leukemia cell line MOLT-4. This cell line is characterized not only by the ability to secrete VEGF, but also for the presence on its surface of the VEGF receptor-1 (VEGFR-1). Previous studies from our laboratory concerned with evaluating early changes in gene expression induced by aplidine in MOLT-4 cells have shown that the drug decreases the expression of VEGFR-1 (Marchini et al. Proc Am Assoc Cancer Res 2000; 41: 833). Here, we report the ability of aplidine to block the VEGF/VEGFR-1 loop. We found that aplidine blocked VEGF secretion that was temporally followed by a decrease in both VEGF and VEGFR-1 production. Aplidine did not directly affect either VEGF transcription or stabilization of its mRNA. Transfection of MOLT-4 cells with an antisense VEGF cDNA construct, resulted in inhibition of colony formations. One clone, transfected with sense VEGF cDNA, secreting 8-10 times more VEGF than parental cells, was less sensitive to aplidine-induced cytotoxicity and apoptosis than control cells. Moreover, addition of VEGF in the medium decreased the activity of aplidine in MOLT-4 cells. These data demonstrate that aplidine inhibits the growth and induces apoptosis in MOLT-4 cells through the inhibition of VEGF secretion which blocks the VEGF/VEGFR-1 autocrine loop necessary for the growth of these cells.


Subject(s)
Antineoplastic Agents/pharmacology , Depsipeptides , Endothelial Growth Factors/antagonists & inhibitors , Leukemia, T-Cell/drug therapy , Lymphokines/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Apoptosis/drug effects , Autocrine Communication , Cell Division/drug effects , DNA Primers/chemistry , Dactinomycin/pharmacology , Electrophoretic Mobility Shift Assay , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Half-Life , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Luciferases/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factors
3.
Oncogene ; 20(50): 7293-300, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11704858

ABSTRACT

Tumor neovascularization is controlled by a balance between positive and negative effectors, whose production can be regulated by oncogenes and tumor suppressor genes. The aim of this study was to investigate whether the angiogenic potential of tumors could also be controlled by p73, a gene homologous to the tumor suppressor p53, whose involvement in tumor angiogenesis is known. We have studied the production of proangiogenic (VEGF, FGF-2, PIGF and PDGF) and antiangiogenic (TSP-1) factors in two p73 overexpressing clones obtained from the human ovarian carcinoma cells A2780. TSP-1 was downregulated in both clones compared to mock transfected cells, both at mRNA and protein level. Conversely, both clones showed an increased production of VEGF mRNA and protein. For both TSP-1 and VEGF, regulation of expression was partially due to modulation of the promoter activity, and was dependent on p53 status. Production of the other angiogenic factors FGF-2, PIGF and PDGF-B was also increased in p73 overexpressing clones. The two clones were more angiogenic than parental cells, as shown in vitro by their increased chemotactic activity for endothelial cells, and in vivo by the generation of more vascularized tumors. These findings suggest a potential role of p73 in tumor angiogenesis.


Subject(s)
DNA-Binding Proteins/physiology , Endothelial Growth Factors/biosynthesis , Lymphokines/biosynthesis , Neovascularization, Pathologic/genetics , Nuclear Proteins/physiology , Thrombospondin 1/biosynthesis , Animals , Blotting, Northern , Carcinoma/pathology , Chemotaxis/drug effects , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genes, p53 , Humans , Lymphokines/genetics , Membrane Proteins , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Ovarian Neoplasms/pathology , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/genetics , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Int J Cancer ; 91(2): 241-7, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11146452

ABSTRACT

Inhibitors of proteases prevent tumor-associated matrix degradation, affecting tumor growth, angiogenesis and metastasis. Our study was designed to investigate the effect of inhibition of matrix metalloproteinases (MMPs) on the growth of experimental hemangiomas, using the model of murine endothelioma eEnd.1 cells. In nude mice, these cells generate hemangiomas, consisting mostly of host-recruited endothelial cells, whose growth requires the activity of MMPs. In vitro, eEnd.1 cells produce factors that recruit endothelial cells and stimulate them to release MMPs. Over-expression of TIMP-2, following retrovirus-mediated gene transfer, decreased tumor growth in vivo. The infected clone CR1, which produces high levels of TIMP-2 (as assessed by Northern blot, ELISA and reverse zymography), formed slow-growing tumors that did not grow beyond 0.4 g, while clone 1H, which produces little TIMP-2, grew not dissimilarly to mock-infected cells and parental e.End.1 cells. Histologically, control tumors presented the features of cavernous hemangiomas, while CR1 tumors had a more solid pattern, showing foci of apoptotic cells. In vitro, TIMP-2 over-expression had no autocrine anti-proliferative effect on endothelioma cells but reduced their ability to recruit endothelial cells. CR1 cells lacked the capacity of mock-infected or parental eEnd.1 cells to stimulate endothelial cell motility and invasiveness. Antibodies against TIMP-2 restored the ability of CR1 to induce endothelial cell invasion. We conclude that, in this model, genetic increase of TIMP-2 inhibits tumor growth, apparently by affecting the recruitment and organization of host endothelial cells by the transformed cells.


Subject(s)
Hemangioma/prevention & control , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Hemangioma/metabolism , Hemangioma/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , RNA, Messenger/analysis , Tissue Inhibitor of Metalloproteinase-2/genetics
5.
Am J Pathol ; 157(5): 1703-11, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073829

ABSTRACT

The endothelial cell-derived endothelin-1 (ET-1) is a potent mitogen for endothelial cells, vascular smooth muscle cells, and tumor cells. In this study, we analyzed the role of ET-1 on human umbilical vein endothelial cell (HUVEC) phenotype related to different stages of angiogenesis. ET-1 promoted HUVEC proliferation, migration, and invasion in a dose-dependent manner. The ET(B) receptor (ET(B)R) antagonist, BQ 788, blocked the angiogenic effects induced by ET-1, whereas the ET(A)R antagonist was less effective. ET-1 stimulated matrix metalloproteinase-2 mRNA expression and metalloproteinase-2 production, as determined by reverse transcriptase-polymerase chain reaction and gelatin zymography. Furthermore ET-1 was able to enhance HUVEC differentiation into cord vascular-like structures on Matrigel. When tested in combination with vascular endothelial growth factor (VEGF), ET-1 enhanced VEGF-induced angiogenic-related effects on endothelial cells in vitro. Finally, using the Matrigel plug neovascularization assay in vivo, ET-1 in combination with VEGF stimulated an angiogenic response comparable to that elicited by basic fibroblast growth factor. These findings demonstrated that ET-1 induces angiogenic responses in cultured endothelial cells through ET(B)R and that stimulates neovascularization in vivo in concert with VEGF. ET-1 and its receptors acting as angiogenic regulators might represent new targets for anti-angiogenic therapy.


Subject(s)
Endothelin-1/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Cell Differentiation , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Drug Synergism , Endothelial Growth Factors/physiology , Endothelium, Vascular/cytology , Humans , Lymphokines/physiology , Matrix Metalloproteinase 2/metabolism , Phenotype , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Exp Cell Res ; 258(2): 384-94, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10896789

ABSTRACT

Matrix metalloproteinases (MMPs) play a critical role in the development of hemangioma-like vascular tumors in mice injected with murine eEnd.1 endothelioma cells. The current study was designed to (a) characterize the presence of MMPs in the vascular tumor, (b) define whether these MMPs originate from the transformed cells or from the recruited stromal cells and (c) study the stimulatory effect of eEnd.1 cells on the production of MMPs by endothelial cells. Several gelatinases were present in the eEnd.1 tumor extract, including latent and activated MMP-2 (72-kDa gelatinase A, EC 3.4.24. 24) and MMP-9 (92-kDa gelatinase B, EC 3.4.24.35). Immunohistochemical analysis of the tumor revealed focal reactivity for MMP-2. No gelatinase was produced by cultured eEnd.1 cells, or by six of nine related endothelioma cell lines, suggesting that stroma cells, particularly endothelial cells recruited by the tumor cells, rather than eEnd.1 cells themselves, are the source of the gelatinases observed in the tumors in vivo. The conditioned medium of eEnd.1 cells stimulated the release of MMP-2 and MMP-1 (interstitial collagenase, EC 3.4.24.7) by endothelial cells, but not of the inhibitor TIMP-2. The increased production of MMP-2 and MMP-1, observed at the protein level (zymogram and Western blot analysis), occurred through a posttranscriptional mechanism, since no increase in mRNA was observed and the stimulation was not prevented by inhibitors of protein synthesis. The inhibitory effects of monensin and brefeldin A, inhibitors of protein secretion, and the decrease in cell-associated MMP-2 in stimulated endothelial cells indicated that regulation occurred mostly at the level of protease secretion. MMPs are known to be regulated at different levels; this study indicates that, in endothelial cells, the stimulation of MMPs can also occur at the level of secretion, a mechanism that provides a rapid mobilization of these crucial enzymes in the early phases of angiogenesis.


Subject(s)
Endothelium, Vascular/enzymology , Hemangioma/enzymology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hemangioma/metabolism , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Mice , Stromal Cells/enzymology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , Tumor Cells, Cultured
7.
Clin Cancer Res ; 5(11): 3603-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589777

ABSTRACT

Matrix metalloproteinases (MMPs) have been implicated in tumor cell invasion, metastasis, and angiogenesis. BAY 12-9566, a novel, non-peptidic biphenyl MMP inhibitor, has shown preclinical activity on a broad range of tumor models and is currently in clinical development. The purpose of this study was to investigate the antiangiogenic activity of BAY 12-9566. In vitro, BAY 12-9566 prevented matrix invasion by endothelial cells in a concentration-dependent manner (IC50 = 8.4x10(-7) M), without affecting cell proliferation. In vivo, oral daily administration of BAY 12-9566 (50-200 mg/kg) inhibited angiogenesis induced by basic fibroblast growth factor in the Matrigel plug assay, reducing the hemoglobin content of the pellets. Histological analysis showed a reduction in the amount of functional vessels within the Matrigel. We conclude that the MMP inhibitor BAY 12-9566 inhibits angiogenesis, a property that further supports its clinical development as an antimetastatic agent.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/pharmacology , Matrix Metalloproteinase Inhibitors , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Organic Chemicals , Animals , Biphenyl Compounds , Cell Division/drug effects , Cells, Cultured , Collagen , Dose-Response Relationship, Drug , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Laminin , Mice , Mice, Inbred C57BL , Phenylbutyrates , Proteoglycans , Umbilical Veins
8.
J Pathol ; 188(1): 76-81, 1999 May.
Article in English | MEDLINE | ID: mdl-10398144

ABSTRACT

Kaposi's sarcoma (KS), a neoplasm often associated with iatrogenic and acquired immunosuppression, is characterized by prominent angiogenesis. Angiogenic factors released by both KS and host cells, as well as HHV-8 and HIV viral products, have been implicated in the pathogenesis of this lesion. Angiogenesis is the result of imbalance among angiogenesis promoters and inhibitors, which disrupts homeostasis. The aim of this study was to investigate the expression and mechanism of KS control of thrombospondin-1 (TSP), a physiological inhibitor of angiogenesis. Immunohistochemical analysis of four KS lesions showed only spotty reactivity for TSP in the stroma and in less than 10 per cent of lesional blood vessels. In addition, the typical KS spindle cells were not stained. In agreement with these findings, decreased levels of TSP were measured with an ELISA assay in the supernatants of cultured KS cells, compared with endothelial cells. In vitro, TSP inhibited the endothelial cell proliferation and motility induced by KS cell supernatants. TSP also prevented endothelial cell motility induced by Tat, a product of HIV-1 endowed with angiogenic potential and implicated in the pathogenesis of AIDS-KS. In vivo, TSP inhibited the angiogenic activity exerted by Tat in the Matrigel sponge model. These results suggest that TSP down-regulation might be permissive for the development of KS-associated angiogenesis.


Subject(s)
Gene Products, tat/pharmacology , Neovascularization, Pathologic/metabolism , Sarcoma, Kaposi/metabolism , Thrombospondin 1/analysis , Animals , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/etiology , Sarcoma, Kaposi/pathology , Thrombospondin 1/pharmacology , Tumor Cells, Cultured
9.
Cell Growth Differ ; 8(4): 471-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9101093

ABSTRACT

Thrombospondin-1 (TSP) inhibits the angiogenic activity of basic fibroblast growth factor (bFGF). Here we address the hypothesis of a direct interaction between TSP and bFGF. Gel permeation chromatography and cross-linking experiments demonstrated that bFGF binds to TSP in solution. bFGF also bound to immobilized TSP in a solid-phase assay. Binding was dose-dependent, with a Kd in the nanomolar range, and was inhibited by anti-TSP antibodies. The 140-kDa carboxyl-terminal fragment of TSP, but not the 25-kDa heparin-binding fragment, fully retained the bFGF binding capacity. Accordingly, binding was inhibited by monoclonal antibodies directed against this fragment. Heparin completely blocked bFGF binding to TSP and to the 140-kDa fragment. TSP and its 140-kDa fragment inhibited the binding of bFGF to endothelial cells at concentrations (> or = 100 nM) that inhibited endothelial cell proliferation but not motility. Low-affinity binding was inhibited more than high-affinity binding (up to 76 and 41% inhibition, respectively), and the inhibition was reversed by anti-TSP antibodies. Vitronectin and transforming growth factor beta, potentially associated with TSP, did not affect bFGF binding to endothelial cells. Although TSP did not affect the activation of the high-affinity receptors, it reduced the long-term internalization of bFGF. We conclude that TSP binds to bFGF through a domain within its 140-kDa fragment, a mechanism that might affect bFGF interaction with endothelial cells, activity, and association with the extracellular matrix.


Subject(s)
Cell Adhesion Molecules/metabolism , Fibroblast Growth Factor 2/metabolism , Membrane Glycoproteins/metabolism , Neovascularization, Physiologic , Animals , Binding Sites , Cattle , Cell Division , Cell Movement , Endothelium, Vascular/cytology , Heparin/metabolism , Molecular Weight , Peptide Fragments/metabolism , Protein Binding , Thrombospondins
10.
Int J Cancer ; 70(1): 72-7, 1997 Jan 06.
Article in English | MEDLINE | ID: mdl-8985093

ABSTRACT

All trans-retinoic acid (ATRA) induces complete remission in acute-promyelocytic-leukemia (APL) patients. This study investigated the adhesive properties of APL cells for the endothelium and the extracellular matrix, their motility and the effect of ATRA on these functions. Blasts from 7 APL patients adhered to resting and IL-1-activated endothelium, to the same degree as normal PMN. Adhesion was partially mediated by ICAM-1 and, for IL-1-activated endothelium, by VCAM-1 and E-selectin. These cells showed less adhesiveness for the matrix than PMN, although they maintained the same substrate preference: they adhered to fibronectin and thrombospondin, but not to laminin and type-IV collagen. Exposure to ATRA in vitro (1 microM for 48 to 96 hr) increased the adhesiveness of APL cells; this effect was particularly evident in the case of sub-endothelial matrix and fibronectin. A similar increment in adhesiveness was observed when comparing cells from 2 patients before and after treatment with ATRA. APL cells migrated in response to fMLP and motility was increased by ATRA. In conclusion, APL cells were less adhesive to the matrix than PMN, but treatment with ATRA considerably enhanced their adhesive properties. This could be important in determining the efflux of leukemic cells from the bone marrow and their tissue infiltration during ATRA therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Leukemia, Promyelocytic, Acute/physiopathology , Tretinoin/pharmacology , Adolescent , Adult , Aged , Child , Child, Preschool , E-Selectin/physiology , Endothelium/physiopathology , Extracellular Matrix , Female , Humans , Intercellular Adhesion Molecule-1/physiology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Male , Middle Aged
11.
Clin Cancer Res ; 2(11): 1843-9, 1996 Nov.
Article in English | MEDLINE | ID: mdl-9816139

ABSTRACT

Endothelial cell migration is a critical event during angiogenesis, and inhibitors of cell motility can affect the angiogenic process. Paclitaxel (Taxol(R)), a microtubule-stabilizing antineoplastic cytotoxic drug, inhibits motility and invasiveness of several cell types. The aim of this study was to investigate the effect of paclitaxel on endothelial cell functions and on angiogenesis. In vivo, paclitaxel (20-28 mg/kg i.v.) significantly inhibited the angiogenic response induced by tumor cell supernatant embedded in a pellet of reconstituted basement membrane (Matrigel) injected s.c. into C57BL/6N mice. In vitro, paclitaxel inhibited endothelial cell proliferation, motility, invasiveness, and cord formation on Matrigel in a dose-dependent manner. The antiangiogenic activity of paclitaxel was not linked to its cytotoxicity, since inhibition of endothelial cell chemotaxis and invasiveness occurred at drug concentrations which did not affect endothelial cell proliferation. Another cytotoxic drug, cisplatin, that inhibited endothelial cell proliferation in vitro, did not affect angiogenesis in vivo. These data indicate that paclitaxel has a strong antiangiogenic activity, a property that might contribute to its antineoplastic activity in vivo.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Microtubules/drug effects , Neovascularization, Pathologic/prevention & control , Paclitaxel/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cells, Cultured , Cisplatin/pharmacology , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Laminin/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Proteoglycans/metabolism
12.
J Natl Cancer Inst ; 87(4): 293-8, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7535861

ABSTRACT

BACKGROUND: The importance of matrix metalloproteinases in angiogenesis, tumor growth, and metastasis is well known. However, little is known about the role of matrix metalloproteinases in the formation of hemangiomas and about the possible therapeutic use of matrix metalloproteinase inhibitors in aggressive vascular tumors. PURPOSE: To study the role of matrix metalloproteinase in vascular tumors, we tested the antineoplastic activity of a synthetic inhibitor of matrix metalloproteinases, batimastat, on an experimental model of hemangioma, formed by murine endothelioma cells transformed by polyoma middle-T oncogene (eEnd.1). METHODS: The effect of batimastat was studied in vivo on the formation of hemorrhaging, cavernous hemangiomas by eEnd.1 endothelioma cells injected subcutaneously in nude mice and on the angiogenic response induced by an endothelioma cell supernatant embedded in a pellet of reconstituted basement membrane (Matrigel). The effect of batimastat was investigated in vitro on endothelial cell proliferation, motility, and invasion of a layer of Matrigel. RESULTS: Daily treatment with batimastat (30, 3, and 0.3 mg/kg at the site of eEnd.1 cell injection) inhibited tumor growth, with increased doubling time. The carboxamide derivative of batimastat, BB-374, a poor inhibitor of matrix metalloproteinase activity, was less active in reducing hemangioma growth. Histologic analysis of treated tumors indicated a reduction in the size of blood-filled spaces and in hemorrhage. Batimastat also inhibited the angiogenic response induced by cultured eEnd.1 endothelioma cell supernatant embedded in a pellet of Matrigel. Batimastat significantly inhibited endothelial cell invasion in vitro through a layer of Matrigel, but it showed no direct cytotoxic activity. CONCLUSIONS: Batimastat reduces in vivo growth of experimental hemangiomas, most probably by blocking endothelial cell recruitment by the transformed cells or by interfering with cell organization in vascular structures. IMPLICATIONS: These results confirm the importance of matrix metalloproteinase in endothelial cell recruitment that occurs in angiogenesis and in the formation of vascular tumors and suggest a therapeutic potential for synthetic matrix metalloproteinase inhibitors.


Subject(s)
Hemangioendothelioma/drug therapy , Metalloendopeptidases/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Phenylalanine/analogs & derivatives , Thiophenes/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Hemangioendothelioma/pathology , Mice , Neoplasm Invasiveness , Phenylalanine/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...