Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Clin Invest ; 115(1): 118-27, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15630451

ABSTRACT

The angiogenic mechanism and therapeutic potential of PDGF-CC, a recently discovered member of the VEGF/PDGF superfamily, remain incompletely characterized. Here we report that PDGF-CC mobilized endothelial progenitor cells in ischemic conditions; induced differentiation of bone marrow cells into ECs; and stimulated migration of ECs. Furthermore, PDGF-CC induced the differentiation of bone marrow cells into smooth muscle cells and stimulated their growth during vessel sprouting. Moreover, delivery of PDGF-CC enhanced postischemic revascularization of the heart and limb. Modulating the activity of PDGF-CC may provide novel opportunities for treating ischemic diseases.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/drug effects , Ischemia/drug therapy , Ischemia/pathology , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/pharmacology , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Hindlimb/blood supply , Hindlimb/drug effects , Humans , Ischemia/chemically induced , Ischemia/metabolism , Lymphokines , Mice , Microcirculation/drug effects , Myocardium/metabolism , Myocardium/pathology , Phosphotyrosine/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Stem Cells/cytology
2.
Oncogene ; 21(3): 427-36, 2002 Jan 17.
Article in English | MEDLINE | ID: mdl-11821955

ABSTRACT

Angiogenesis is a complex biological process involving the coordinated modulation of many genes. Histone deacetylases (HDAC) are a growing family of enzymes that mediate the availability of chromatin to the transcriptional machinery. Trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA), two HDAC inhibitors known to relieve gene silencing, were evaluated as potential antiangiogenic agents. TSA and SAHA were shown to prevent vascular endothelial growth factor (VEGF)-stimulated human umbilical cord endothelial cells (HUVEC) from invading a type I collagen gel and forming capillary-like structures. SAHA and TSA inhibited the VEGF-induced formation of a CD31-positive capillary-like network in embryoid bodies and inhibited the VEGF-induced angiogenesis in the CAM assay. TSA also prevented, in a dose-response relationship, the sprouting of capillaries from rat aortic rings. TSA inhibited in a dose-dependent and reversible fashion the VEGF-induced expression of VEGF receptors, VEGFR1, VEGFR2, and neuropilin-1. TSA and SAHA upregulated the expression by HUVEC of semaphorin III, a recently described VEGF competitor, at both mRNA and protein levels. This effect was specific to endothelial cells and was not observed in human fibroblasts neither in vascular smooth muscle cells. These observations provide a conspicuous demonstration that HDAC inhibitors are potent anti-angiogenic factors altering VEGF signaling.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Growth Factors/pharmacology , Histone Deacetylase Inhibitors , Lymphokines/pharmacology , Semaphorin-3A , Signal Transduction/drug effects , Animals , Aorta/drug effects , Blotting, Western , Carrier Proteins/genetics , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal, 28S/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vorinostat
3.
FASEB J ; 18(3): 565-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14734647

ABSTRACT

To generate doxorubicin (Dox) specifically at the tumor site, the chemotherapeutic agent was incorporated into a prodrug by linkage to a peptide specifically recognized by plasmin, which is overproduced in many cancers. ST-9905, which contains an elongated self-elimination spacer, is activated more rapidly in vitro by plasmin than is ST-9802. Prodrug activation in vitro depended on the level of urokinase produced by tumor cells and was inhibited by aprotinin, a plasmin inhibitor. Comparison of equimolar concentrations of ST-9905, ST-9802, and Dox in EF43.fgf-4 and MCF7 models revealed that both prodrugs, in sharp contrast to Dox, displayed antiproliferative and antiangiogenic activities without discernible toxicity. Although MCF7 cells are poor urokinase producers in vitro, prodrug efficacy in this model may be explained by production of plasmin by tumor-infiltrating host cells. Mice treated with equitoxic concentrations (maximum tolerated doses) of prodrugs showed 100% survival and negligible body weight loss, in contrast to results after Dox treatment. ST-9905 was substantially more effective than ST-9802 and induced similar tumor growth inhibition as Dox but without apparent toxicity. This finding may be explained by the elongated spacer, which facilitates enzymatic prodrug activation. These data validate both the use of elongated spacers in vivo and the concept of targeting anticancer prodrugs to tumor-associated plasmin.


Subject(s)
Adenocarcinoma/drug therapy , Doxorubicin/pharmacology , Fibrinolysin/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Prodrugs/pharmacology , Adenocarcinoma/blood supply , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Biotransformation , Body Weight/drug effects , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor/transplantation , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/therapeutic use , Doxorubicin/toxicity , Female , Humans , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasm Proteins/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Prodrugs/toxicity , Urokinase-Type Plasminogen Activator/metabolism , Xenograft Model Antitumor Assays
4.
FASEB J ; 16(13): 1802-4, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12354694

ABSTRACT

Endostatin and angiostatin are known as tumor-derived angiogenesis inhibitors, but their mechanisms of action are not yet completely defined. We report here that endostatin and angiostatin, delivered by adenoviral vectors, reduced in vitro the neovessel formation in the mouse aortic ring assay by 85 and 40%, respectively. We also demonstrated in vivo that both endostatin and angiostatin inhibited local invasion and tumor vascularization of transplanted murine malignant keratinocytes, and reduced by 50 and 90% the development of highly vascularized murine mammary tumors. This inhibition of tumor growth was associated with a reduction of tumor vascularization. Expression analysis of vascular endothelial growth factor (VEGF) carried out in the mouse aortic ring model revealed a 3- to 10-fold down-regulation of VEGF mRNA expression in endostatin-treated rings. A similar down-regulation of VEGF expression at both mRNA and protein levels was also observed in the two in vivo cancer models after treatment with each angiogenesis inhibitor. This suggests that endostatin and angiostatin effects may be mediated, at least in part, by their ability to down-regulate VEGF expression within the tumor. This work provides evidence that endostatin and angiostatin act on tumor cells themselves.


Subject(s)
Collagen/physiology , Endothelial Growth Factors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Peptide Fragments/physiology , Plasminogen/physiology , Adenoviridae/genetics , Angiostatins , Animals , Aorta, Thoracic/metabolism , Blood Vessels/growth & development , Blotting, Western , Collagen/genetics , Culture Techniques , Down-Regulation , Endostatins , Endothelial Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Peptide Fragments/genetics , Plasminogen/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
FASEB J ; 16(2): 147-54, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11818362

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1) is believed to control proteolytic activity and cell migration during angiogenesis. We previously demonstrated in vivo that this inhibitor is necessary for optimal tumor invasion and vascularization. We also showed that PAI-1 angiogenic activity is associated with its control of plasminogen activation but not with the regulation of cell-matrix interaction. To dissect the role of the various components of the plasminogen activation system during angiogenesis, we have adapted the aortic ring assay to use vessels from gene-inactivated mice. The single deficiency of tPA, uPA, or uPAR, as well as combined deficiencies of uPA and tPA, did not dramatically affect microvessel formation. Deficiency of plasminogen delayed microvessel outgrowth. Lack of PAI-1 completely abolished angiogenesis, demonstrating its importance in the control of plasmin-mediated proteolysis. Microvessel outgrowth from PAI-1-/- aortic rings could be restored by adding exogenous PAI-1 (wild-type serum or purified recombinant PAI-1). Addition of recombinant PAI-1 led to a bell-shaped angiogenic response clearly showing that PAI-1 is proangiogenic at physiological concentrations and antiangiogenic at higher levels. Using specific PAI-1 mutants, we could demonstrate that PAI-1 promotes angiogenesis at physiological (nanomolar) concentrations through its antiproteolytic activity rather than by interacting with vitronectin.


Subject(s)
Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Aorta/drug effects , Aorta/growth & development , Blood Vessels/drug effects , Blood Vessels/growth & development , Culture Techniques , Dose-Response Relationship, Drug , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Plasminogen/genetics , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/physiology , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/genetics
6.
Clin Cancer Res ; 10(12 Pt 1): 4038-47, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15217936

ABSTRACT

PURPOSE: The implication of matrix metalloproteinases (MMPs) in the major stages of cancer progression has fueled interest in the design of synthetic MMP inhibitors (MMPIs) as a novel anticancer therapy. Thus far, drugs used in clinical trials are broad-spectrum MMPIs the therapeutic index of which proved disappointingly low. The development of selective MMPIs for tumor progression-associated MMPs is, thus, likely to offer improved therapeutic possibilities. EXPERIMENTAL DESIGN: The anti-invasive capacity of a series of pyrimidine-trione derivatives was tested in vitro in a chemoinvasion assay, and the most potent compound was further evaluated in vivo in different human tumor xenograft models. The activity of this novel selective MMPI was compared with BB-94, a broad-spectrum inhibitor. RESULTS: Ro-28-2653, an inhibitor with high selectivity for MMP-2, MMP-9, and membrane type 1 (MT1)-MMP, showed the highest anti-invasive activity in vitro. In vivo, Ro-28-2653 reduced the growth of tumors induced by the inoculation of different cell lines producing MMPs and inhibited the tumor-promoting effect of fibroblasts on breast adenocarcinoma cells. Furthermore, Ro-28-2653 reduced tumor vascularization and blocked angiogenesis in a rat aortic ring assay. In contrast, BB-94 up-regulated MMP-9 expression in tumor cells and promoted angiogenesis in the aortic ring assay. CONCLUSION: Ro-28-2653, a selective and orally bioavailable MMPI with inhibitory activity against MMPs expressed by tumor and/or stromal cells, is a potent antitumor and antiangiogenic agent. In contrast to broad-spectrum inhibitors, the administration of Ro-28-2653 was not associated with the occurrence of adverse side effects that might hamper the therapeutic potential of these drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Matrix Metalloproteinase Inhibitors , Phenylalanine/analogs & derivatives , Piperazines/pharmacology , Pyrimidines/pharmacology , Adenocarcinoma/drug therapy , Administration, Oral , Animals , Aorta/pathology , Breast Neoplasms/drug therapy , Cell Line, Tumor , DNA, Complementary/metabolism , Disease Progression , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Humans , Inhibitory Concentration 50 , Matrix Metalloproteinase 2/metabolism , Microscopy, Fluorescence , Models, Chemical , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Rats , Thiophenes/pharmacology , Time Factors , Up-Regulation
7.
J Natl Cancer Inst ; 107(4)2015 Apr.
Article in English | MEDLINE | ID: mdl-25710962

ABSTRACT

BACKGROUND: Matrix metalloproteinase (MMP) 14 may mediate tumor progression through vascular and immune-modulatory effects. METHODS: Orthotopic murine breast tumors (4T1 and E0771 with high and low MMP14 expression, respectively; n = 5-10 per group) were treated with an anti-MMP14 inhibitory antibody (DX-2400), IgG control, fractionated radiation therapy, or their combination. We assessed primary tumor growth, transforming growth factor ß (TGFß) and inducible nitric oxide synthase (iNOS) expression, macrophage phenotype, and vascular parameters. A linear mixed model with repeated observations, with Mann-Whitney or analysis of variance with Bonferroni post hoc adjustment, was used to determine statistical significance. All statistical tests were two-sided. RESULTS: DX-2400 inhibited tumor growth compared with IgG control treatment, increased macrophage numbers, and shifted the macrophage phenotype towards antitumor M1-like. These effects were associated with a reduction in active TGFß and SMAD2/3 signaling. DX-2400 also transiently increased iNOS expression and tumor perfusion, reduced tissue hypoxia (median % area: control, 20.2%, interquartile range (IQR) = 6.4%-38.9%; DX-2400: 1.2%, IQR = 0.2%-3.2%, P = .044), and synergistically enhanced radiation therapy (days to grow to 800mm(3): control, 12 days, IQR = 9-13 days; DX-2400 plus radiation, 29 days, IQR = 26-30 days, P < .001) in the 4T1 model. The selective iNOS inhibitor, 1400W, abolished the effects of DX-2400 on vessel perfusion and radiotherapy. On the other hand, DX-2400 was not capable of inducing iNOS expression or synergizing with radiation in E0771 tumors. CONCLUSION: MMP14 blockade decreased immunosuppressive TGFß, polarized macrophages to an antitumor phenotype, increased iNOS, and improved tumor perfusion, resulting in reduced primary tumor growth and enhanced response to radiation therapy, especially in high MMP14-expressing tumors.


Subject(s)
Amidines/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Benzylamines/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Macrophages/drug effects , Matrix Metalloproteinase 14/drug effects , Matrix Metalloproteinase 14/metabolism , Nitric Oxide Synthase Type II/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin G/blood , Macrophages/enzymology , Mammary Neoplasms, Experimental , Mice , Neovascularization, Pathologic , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phenotype , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
8.
Biol Proced Online ; 4: 24-31, 2002 Oct 28.
Article in English | MEDLINE | ID: mdl-12734572

ABSTRACT

Angiogenesis, a key step in many physiological and pathological processes, involves proteolysis of the extracellular matrix. To study the role of two enzymatic families, serine-proteases and matrix metalloproteases in angiogenesis, we have adapted to the mouse, the aortic ring assay initially developed in the rat. The use of deficient mice allowed us to demonstrate that PAI-1 is essential for angiogenesis while the absence of an MMP, MMP-11, did not affect vessel sprouting. We report here that this model is attractive to elucidate the cellular and molecular mechanisms of angiogenesis, to identify, characterise or screen "pro- or anti-angiogenic agents that could be used for the treatment of angiogenesis-dependent diseases. Approaches include using recombinant proteins, synthetic molecules and adenovirus-mediated gene transfer.

9.
Invest Ophthalmol Vis Sci ; 44(7): 3186-93, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12824270

ABSTRACT

PURPOSE: VEGF has been shown to be necessary, but not sufficient alone, for the development of subretinal pathologic angiogenesis. In the current study, the influence of placental growth factor (PlGF), a member of the VEGF family, in human and experimental choroidal neovascularization (CNV) was investigated. METHODS: The presence of VEGF family member mRNA was evaluated by RT-PCR in neovascular membranes extracted during surgery. The spatial and temporal pattern of VEGF isoforms and PlGF mRNA expression were explored by using the laser capture catapulting technique and RT-PCR in a murine laser-induced model and in vitro. PlGF expression was also studied in human donor eyes. The influence of endogenous PlGF was evaluated in deficient mice (PlGF(-/-)) and by antibody-mediated neutralization of the PlGF receptor. RESULTS: Human neovascular membranes consistently expressed VEGF-A, -B, and -C; PlGF; and VEGFR-1 and -2. The VEGF(120) isoform mRNA was primarily induced in early stages of angiogenesis in vivo and in vitro. PlGF mRNA expression was present in the intact choroid and significantly upregulated during the course of experimental CNV. Both deficient PlGF expression in PlGF(-/-) mice and PlGF receptor neutralization in wild-type mice prevented the development of choroidal neovascularization induced by laser. CONCLUSIONS: These observations demonstrate the participation of PlGF in experimental CNV. They identify therefore PlGF as an additional promising target for ocular antiangiogenic strategies.


Subject(s)
Angiogenesis Inducing Agents/genetics , Choroidal Neovascularization/metabolism , Pregnancy Proteins/genetics , Angiogenesis Inducing Agents/metabolism , Animals , Animals, Genetically Modified , Choroidal Neovascularization/pathology , Disease Models, Animal , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Female , Fluorescent Antibody Technique, Indirect , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Male , Mice , Placenta Growth Factor , Pregnancy Proteins/metabolism , Protein Isoforms , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factors
10.
Biochem Pharmacol ; 63(2): 179-89, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11841792

ABSTRACT

In myeloid leukemia, immature leukemic cells are able to egress into peripheral blood to infiltrate extra-medullary organs. We therefore analyzed the migrating and invasive potential of human HL-60 and NB4 cell lines, representative of acute myelogenous leukemia, their ability to express matrix metalloproteases (MMPs), tissue inhibitors of metalloproteases (TIMPs) and urokinase plasminogen activator (uPA) in response to differentiating agents. Granulocytic differentiation by all-trans-retinoic acid (ATRA) and aclacinomycin (ACLA) strongly increased HL-60 and NB4 cell migration and invasion. At mRNA and protein levels, these cell lines produced significant amounts of MMP-9 (HL-60

Subject(s)
Aclarubicin/analogs & derivatives , Aclarubicin/pharmacology , Cell Differentiation/drug effects , Leukemia/enzymology , Phenylalanine/analogs & derivatives , Serine Endopeptidases/biosynthesis , Tretinoin/pharmacology , Antineoplastic Agents/pharmacology , Cell Differentiation/physiology , Cell Movement/drug effects , HL-60 Cells , Humans , Leukemia/metabolism , Leukemia/pathology , Matrix Metalloproteinase 9/biosynthesis , Neoplasm Invasiveness , Phenylalanine/pharmacology , Thiophenes/pharmacology , Tissue Inhibitor of Metalloproteinases/biosynthesis , Tumor Cells, Cultured
11.
Biochem Res Int ; 2011: 191670, 2011.
Article in English | MEDLINE | ID: mdl-21152183

ABSTRACT

MMP intervention strategies have met with limited clinical success due to severe toxicities. In particular, treatment with broad-spectrum MMP-inhibitors (MMPIs) caused musculoskeletal pain and inflammation. Selectivity may be essential for realizing the clinical potential of MMPIs. Here we review discoveries pinpointing membrane-bound MMPs as mediators of mechanisms underlying cancer and inflammation and as possible therapeutic targets for prevention/treatment of these diseases. We discuss strategies to target these therapeutic proteases using highly selective inhibitory agents (i.e., human blocking antibodies) against individual membrane-bound MMPs.

12.
Cancer Res ; 69(4): 1517-26, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19208838

ABSTRACT

Inhibition of specific matrix metalloproteinases (MMP) is an attractive noncytotoxic approach to cancer therapy. MMP-14, a membrane-bound zinc endopeptidase, has been proposed to play a central role in tumor growth, invasion, and neovascularization. Besides cleaving matrix proteins, MMP-14 activates proMMP-2 leading to an amplification of pericellular proteolytic activity. To examine the contribution of MMP-14 to tumor growth and angiogenesis, we used DX-2400, a highly selective fully human MMP-14 inhibitory antibody discovered using phage display technology. DX-2400 blocked proMMP-2 processing on tumor and endothelial cells, inhibited angiogenesis, and slowed tumor progression and formation of metastatic lesions. The combination of potency, selectivity, and robust in vivo activity shows the potential of a selective MMP-14 inhibitor for the treatment of solid tumors.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Enzyme Inhibitors/therapeutic use , Matrix Metalloproteinase Inhibitors , Neovascularization, Pathologic/prevention & control , Animals , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Genes, Reporter , Humans , Immunohistochemistry , Mice , Neoplasm Invasiveness/pathology , Transfection , Transplantation, Heterologous , Umbilical Veins/cytology , Umbilical Veins/drug effects
13.
Neoplasia ; 9(11): 927-37, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18030361

ABSTRACT

Novel inhibitors of the urokinase-mediated plasminogen (plg) activation system are potentially of great clinical benefit as anticancer treatments. Using phage display, we identified DX-1000 a tissue factor pathway inhibitor-derived Kunitz domain protein which is a specific high-affinity inhibitor of plasmin (pln) (K(i) = 99 pM). When tested in vitro, DX-1000 blocks plasmin-mediated pro-matrix metalloproteinase-9 (proMMP-9) activation on cells and dose-dependently inhibits tube formation, while not significantly affecting hemostasis and coagulation. However, this low-molecular weight protein inhibitor ( approximately 7 kDa) exhibits rapid plasma clearance in mice and rabbits, limiting its potential clinical use in chronic diseases. After site-specific PEGylation, DX-1000 retains its activity and exhibits a decreased plasma clearance. This PEGylated derivative is effective in vitro, as well as potent in inhibiting tumor growth of green fluorescent protein (GFP)-labeled MDA-MB-231 cells. 4PEG-DX-1000 treatment causes a significant reduction of urokinase-type plasminogen activator (uPA) and plasminogen expressions, a reduction of tumor proliferation, and vascularization. 4PEG-DX-1000 treatment significantly decreases the level of active mitogen-activated protein kinase (MAPK) in the primary tumors and reduces metastasis incidence. Together, our results demonstrate the potential value of plasmin inhibitors as therapeutic agents for blocking breast cancer growth and metastasis.


Subject(s)
Antifibrinolytic Agents/pharmacology , Antineoplastic Agents/pharmacology , Polyethylene Glycols/pharmacology , Animals , Antifibrinolytic Agents/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Blood Coagulation/drug effects , Cell Line , Dose-Response Relationship, Drug , Enzyme Precursors/antagonists & inhibitors , Female , Hemostasis/drug effects , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 9 , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Rabbits , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology
14.
J Pharmacol Exp Ther ; 302(3): 1055-61, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183663

ABSTRACT

Bisphosphonate drugs inhibit osteoclastic bone resorption and are widely used to treat skeletal complications in patients with tumor-induced osteolysis. We now show that zoledronic acid, a new generation bisphosphonate with a heterocyclic imidazole substituent, is also a potent inhibitor of angiogenesis. In vitro, zoledronic acid inhibits proliferation of human endothelial cells stimulated with fetal calf serum, basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (IC(50) values 4.1, 4.2, and 6.9 microM, respectively), and modulates endothelial cell adhesion and migration. In cultured aortic rings and in the chicken egg chorioallantoic membrane assay, zoledronic acid reduces vessel sprouting. When administered systemically to mice, zoledronic acid potently inhibits the angiogenesis induced by subcutaneous implants impregnated with bFGF [ED(50), 3 microg/kg (7.5 nmol/kg) s.c.]. These findings indicate that zoledronic acid has marked antiangiogenic properties that could augment its efficacy in the treatment of malignant bone disease and extend its potential clinical use to other diseases with an angiogenic component.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Chorion/chemistry , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Growth Substances/administration & dosage , Growth Substances/pharmacology , Humans , Image Cytometry , Lymphokines/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Pamidronate , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Zoledronic Acid
15.
Am J Pathol ; 165(3): 869-78, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331411

ABSTRACT

Matrix metalloproteinases (MMPs) are known to play a role in cell growth, invasion, angiogenesis, metastasis, and bone degradation, all important events in the pathogenesis of cancer. Multiple myeloma is a B-cell cancer characterized by the proliferation of malignant plasma cells in the bone marrow, increased angiogenesis, and the development of osteolytic bone disease. The role of MMPs in the development of multiple myeloma is poorly understood. Using SC-964, a potent inhibitor of several MMPs (MMP-2, -3, -8, -9, and -13), we investigated the role of MMPs in the 5T2MM murine model. Reverse transcriptase-polymerase chain reaction demonstrated the presence of mRNA for MMP-2, -8, -9, and -13 in 5T2MM-diseased bone marrow. Mice bearing 5T2MM cells were given access to food containing SC-964. The concentration of SC-964 measured in the plasma of mice after 11 days of treatment was able to inhibit MMP-9 activity in gelatin zymography. Treatment of 5T2MM-bearing mice resulted in a significant reduction in tumor burden, a significant decrease in angiogenesis, and partially protective effect against the development of osteolytic bone disease. The direct role of MMPs in these different processes was confirmed by in vitro experiments. All these results support the multifunctional role of MMPs in the development of multiple myeloma.


Subject(s)
Bone Diseases/prevention & control , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Matrix Metalloproteinases/physiology , Multiple Myeloma/enzymology , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Bone Diseases/enzymology , Cell Division/drug effects , Diet , Female , Mice , Mice, Inbred C57BL , Multiple Myeloma/blood supply , Multiple Myeloma/prevention & control , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/prevention & control , Osteolysis/enzymology , Osteolysis/prevention & control , Plasma Cells/metabolism , Plasma Cells/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL