Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer J ; 7(5): 427-36, 2001.
Article in English | MEDLINE | ID: mdl-11693902

ABSTRACT

Cancer chemotherapy utilized at maximum tolerated, toxic doses, rarely results in sustained total tumor eradication, with patients ultimately failing a variety of chemotherapeutic regimens. If tumors respond, the constituent cancer cells acquire resistance to chemotherapeutic agents, largely due to tremendous genetic instability. Changing the target of these agents to the tumor's proliferating microvasculature, a more genetically stable cell, may provide important therapeutic advantages. Modifying the cyclic schedule and high doses of chemotherapeutic agents to a continuous, lower dose, "metronomic" regimen increases the efficacy of targeting the tumor microvasculature, and produces therapeutic activity with decreased toxicity. Preclinically, the antiangiogenic properties of continuous, lower-dose chemotherapy can be further enhanced by the concurrent administration of a selective angiogenesis inhibitor. Because the target is not exclusively the tumor cell, this antiangiogenic combination strategy provides the opportunity to delay or possibly avoid acquired resistance. Preclinical and clinical observations provide a basis for treating cancer as a chronic disease, using protracted, continuous dosing. Because appropriate chemotherapeutic agents and commercially available compounds that inhibit angiogenesis are readily available, it would be beneficial to initiate clinical trials to evaluate the antiangiogenic scheduling of chemotherapy expeditiously.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Drug Resistance, Neoplasm , Forecasting , Humans , Mice
2.
Cancer Metastasis Rev ; 19(1-2): 19-27, 2000.
Article in English | MEDLINE | ID: mdl-11191059

ABSTRACT

Cyclooxygenase-2 (COX-2) is an immediate early response gene that can be induced by a variety of tumor promoters, cytokines, growth factors and hypoxia. COX-2 overexpression is linked to all stages of carcinogenesis with the enzyme localized to the neoplastic cells, microvascular endothelial cells, and stromal fibroblasts. The contributions of COX-2 in tumor angiogenesis include: (a) the increased expression of the proangiogenic growth factor VEGF; (b) the production of the eicosanoid products thromboxane A2, PGE2 and PGI2 that can directly stimulate endothelial cell migration and growth factor-induced angiogenesis; and potentially, (c) the inhibition of endothelial cell apoptosis by stimulation of Bcl-2 or Akt activation. Selective pharmacological inhibitors of COX-2 as angiosuppressive agents could have therapeutic benefit in the treatment of neoplastic disease from prevention through treatment of advanced metastatic disease. These agents are safe and well tolerated and can be added to chemotherapy and radiation therapy where angiogenesis inhibitors appear to provide at least additive therapeutic benefit.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Endothelial Growth Factors/genetics , Endothelial Growth Factors/physiology , Endothelium, Vascular/physiopathology , Humans , Isoenzymes/genetics , Lymphokines/genetics , Lymphokines/physiology , Membrane Proteins , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Prostaglandin-Endoperoxide Synthases/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Biochem J ; 340 ( Pt 1): 77-84, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10229661

ABSTRACT

Angiostatin, a kringle-containing fragment of plasminogen, is a potent inhibitor of angiogenesis. The mechanism(s) responsible for the anti-angiogenic properties of angiostatin are unknown. We now report that human angiostatin blocks plasmin(ogen)-enhanced in vitro invasion of tissue plasminogen activator (t-PA)-producing endothelial and melanoma cells. Kinetic analyses demonstrated that angiostatin functions as a non-competitive inhibitor of extracellular-matrix (ECM)-enhanced, t-PA-catalysed plasminogen activation, with a Ki of 0.9+/-0.03 microM. This mechanism suggests that t-PA has a binding site for the inhibitor angiostatin, as well as for its substrate plasminogen that, when occupied, prevents ternary complex formation between t-PA, plasminogen and matrix protein. Direct binding experiments confirmed that angiostatin bound to t-PA with an apparent Kd [Kd(app)] of 6.7+/-0.7 nM, but did not bind with high affinity to ECM proteins. Together, these data suggest that angiostatin in the cellular micro-environment can inhibit matrix-enhanced plasminogen activation, resulting in reduced invasive activity, and suggest a biochemical mechanism whereby angiostatin-mediated regulation of plasmin formation could influence cellular migration and invasion.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Extracellular Matrix/physiology , Melanoma/pathology , Peptide Fragments/pharmacology , Plasminogen/metabolism , Plasminogen/pharmacology , Angiostatins , Animals , Antibodies/pharmacology , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Extracellular Matrix Proteins/metabolism , Female , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Humans , Kinetics , Melanoma/enzymology , Melanoma/metabolism , Mice , Neovascularization, Pathologic , Peptide Fragments/metabolism , Protein Binding , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/immunology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , Tumor Cells, Cultured , alpha-2-Antiplasmin/metabolism , alpha-2-Antiplasmin/pharmacology
4.
Cancer Res ; 58(24): 5686-9, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9865723

ABSTRACT

Angiostatin, a proteolytic fragment of plasminogen, inhibits the growth of primary and metastatic tumors by suppressing angiogenesis. When used in combination with ionizing radiation (IR), angiostatin demonstrates potent antitumor synergism, largely caused by inhibition of the tumor microvasculature. We report here the temporal interaction of angiostatin and IR in Lewis lung carcinoma (LLC) tumors growing in the hind limbs of syngeneic mice. Tumors with an initial mean volume of 510 +/- 151 mm3 were treated with IR alone (20 Gy x 2 doses on days 0 and 1), angiostatin alone (25 mg/kg/day divided twice daily) on days 0 through 13, or a combination of the two as follows: (a) IR plus angiostatin (days 0 through 13); (b) IR plus angiostatin (days 0 and 1); and (c) IR followed by angiostatin beginning on the day after IR completion and given daily thereafter (days 2 through 13). By day 14, tumors in untreated control mice had grown to 6110 +/- 582 mm3, whereas in mice treated with: (a) IR alone, tumors had grown to 2854 +/- 338 mm3 (P < 0.05 compared with untreated controls); and (b) angiostatin alone, tumors had grown to 3666 +/- 453 mm3 (P < 0.05 compared with untreated controls). In combined-treatment groups, in mice treated with: (a) IR plus longer-course angiostatin, tumors reached 2022 +/- 282 mm3 (P = 0.036 compared with IR alone); (b) IR followed by angiostatin, tumors reached 2677 +/- 469 mm3 (P > 0.05 compared with IR alone); and (c) IR plus short-course angiostatin, tumors reached 1032 +/- 78 mm3 (P < 0.001 compared with IR alone). These findings demonstrate that the efficacy of experimental radiation therapy is potentiated by brief concomitant exposure of the tumor vasculature to angiostatin.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Angiostatins , Animals , Combined Modality Therapy , Drug Administration Schedule , Female , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Plasminogen/administration & dosage , Time Factors , Tumor Cells, Cultured
5.
Nature ; 394(6690): 287-91, 1998 Jul 16.
Article in English | MEDLINE | ID: mdl-9685160

ABSTRACT

Angiogenesis, the formation of new capillaries from pre-existing vessels, is essential for tumour progression. Angiostatin, a proteolytic fragment of plasminogen that was first isolated from the serum and urine of tumour-bearing mice, inhibits angiogenesis and thereby growth of primary and metastatic tumours. Radiotherapy is important in the treatment of many human cancers, but is often unsuccessful because of tumour cell radiation resistance. Here we combine radiation with angiostatin to target tumour vasculature that is genetically stable and therefore less likely to develop resistance. The results show an antitumour interaction between ionizing radiation and angiostatin for four distinct tumour types, at doses of radiation that are used in radiotherapy. The combination produced no increase in toxicity towards normal tissue. In vitro studies show that radiation and angiostatin have combined cytotoxic effects on endothelial cells, but not tumour cells. In vivo studies show that these agents, in combination, target the tumour vasculature. Our results provide support for combining ionizing radiation with angiostatin to improve tumour eradication without increasing deleterious effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Angiostatins , Animals , Apoptosis , Carcinoma, Lewis Lung/blood supply , Combined Modality Therapy , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured
6.
Cancer Res ; 57(23): 5277-80, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9393749

ABSTRACT

Angiostatin inhibits angiogenesis and metastatic tumor growth; however, its usefulness in treating primary nonmetastasizing tumors is less well understood. We now report the effectiveness of human angiostatin administration in a mouse hemangioendothelioma model. Human angiostatin was administered to mice with s.c. hemangioendothelioma and associated disseminated intravascular coagulopathy (Kasabach-Merritt syndrome). Angiostatin significantly reduced tumor volume in comparison to nontreated controls, increased survival, and prevented the profound thrombocytopenia and anemia of Kasabach-Merritt syndrome. Apoptosis of tumor cells was induced by angiostatin, but tumor cell proliferation was not inhibited. These data suggest angiostatin as a novel treatment for nonmetastasizing vascular tumors and for Kasabach-Merritt syndrome.


Subject(s)
Anemia/prevention & control , Antineoplastic Agents/therapeutic use , Hemangioendothelioma/drug therapy , Hemangioendothelioma/pathology , Hemangioma/prevention & control , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Thrombocytopenia/prevention & control , Angiostatins , Animals , Apoptosis/drug effects , Cell Division/drug effects , Female , Humans , Mice , Mice, Nude , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Spleen/drug effects , Spleen/pathology , Syndrome
7.
J Clin Invest ; 100(10): 2580-7, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9366573

ABSTRACT

A key component of the nitric oxide-cyclic guanosine monophosphate (cGMP) pathway in smooth muscle cells (SMC) is the type I GMP-dependent protein kinase (PK-G I). Activation of PK-G I mediates the reduction of cytoplasmic calcium concentrations and vasorelaxation. In this manuscript, we demonstrate that continuous exposure of SMC in culture to the nitrovasodilators S-nitroso-N-acetylpenicillamine (SNAP) or sodium nitroprusside (SNP) results in approximately 75% suppression of PK-G I mRNA by 48 h. PK-G I mRNA and protein were also suppressed by continuous exposure to cGMP analogues 8-bromo- and 8-(4-chlorophenylthio) guanosine-3,5-monophosphate or the cAMP analogue dibutyryl cAMP. These results suggest that activation of one or both of the cyclic nucleotide-dependent protein kinases mediates PK-G I mRNA suppression. Using isoform-specific cDNA probes, only the PK-G I alpha was detected in SMC, either at baseline or after suppression, while PK-G I beta was not detected, indicating that isoform switch was not contributing to the gene regulation. Using the transcription inhibitor actinomycin D, the PK-G I mRNA half-life in bovine SMC was observed to be 5 h. The half-life was not affected by the addition of SNAP to actinomycin D, indicating no effect on PK-G I mRNA stability. Nuclear runoff studies indicated a suppression of PK-G I gene transcription by SNAP. PK-G I suppression was also observed in vivo in rats given isosorbide dinitrate in the drinking water, with a dose-dependent suppression of PK-G I protein in the aorta. PK-G I antigen in whole rat lung extract was also suppressed by administration of isosorbide or theophylline in the drinking water. These data may contribute to our understanding of nitrovasodilator resistance, a phenomenon resulting from continuous exposure to nitroglycerin or other nitrovasodilators.


Subject(s)
Cyclic AMP/physiology , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Cyclic GMP/physiology , Gene Expression Regulation, Enzymologic/drug effects , Muscle, Smooth, Vascular/enzymology , Transcription, Genetic/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Bucladesine/pharmacology , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Colforsin/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Isosorbide/pharmacology , Kinetics , Male , Muscle, Smooth, Vascular/drug effects , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , RNA, Messenger/biosynthesis , Rats , S-Nitroso-N-Acetylpenicillamine , Theophylline/pharmacology , Thionucleotides/pharmacology
8.
Proc Natl Acad Sci U S A ; 94(20): 10868-72, 1997 Sep 30.
Article in English | MEDLINE | ID: mdl-9380726

ABSTRACT

Angiostatin, a potent naturally occurring inhibitor of angiogenesis and growth of tumor metastases, is generated by cancer-mediated proteolysis of plasminogen. Human prostate carcinoma cells (PC-3) release enzymatic activity that converts plasminogen to angiostatin. We have now identified two components released by PC-3 cells, urokinase (uPA) and free sulfhydryl donors (FSDs), that are sufficient for angiostatin generation. Furthermore, in a defined cell-free system, plasminogen activators [uPA, tissue-type plasminogen activator (tPA), or streptokinase], in combination with one of a series of FSDs (N-acetyl-L-cysteine, D-penicillamine, captopril, L-cysteine, or reduced glutathione] generate angiostatin from plasminogen. An essential role of plasmin catalytic activity for angiostatin generation was identified by using recombinant mutant plasminogens as substrates. The wild-type recombinant plasminogen was converted to angiostatin in the setting of uPA/FSD; however, a plasminogen activation site mutant and a catalytically inactive mutant failed to generate angiostatin. Cell-free derived angiostatin inhibited angiogenesis in vitro and in vivo and suppressed the growth of Lewis lung carcinoma metastases. These findings define a direct mechanism for cancer-cell-mediated angiostatin generation and permit large-scale production of bioactive angiostatin for investigation and potential therapeutic application.


Subject(s)
Antineoplastic Agents/metabolism , Neovascularization, Pathologic/prevention & control , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Plasminogen/metabolism , Prostatic Neoplasms/metabolism , Angiostatins , Animals , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell-Free System , Chromatography, Affinity , Humans , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/isolation & purification , Plasminogen/isolation & purification , Prostatic Neoplasms/pathology
9.
Cancer Res ; 56(21): 4887-90, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8895739

ABSTRACT

Angiostatin is an inhibitor of angiogenesis and metastatic growth that is found in tumor-bearing animals and can be generated in vitro by the proteolytic cleavage of plasminogen. The mechanism by which angiostatin is produced in vivo has not been defined. We now demonstrate that human prostate carcinoma cell lines (PC-3, DU-145, and LN-CaP) express enzymatic activity that can generate bioactive angiostatin from purified human plasminogen or plasmin. Affinity purified PC-3-derived angiostatin inhibited human endothelial cell proliferation, basic fibroblast growth factor-induced migration, endothelial cell tube formation, and basic fibroblast growth factor-induced corneal angiogenesis. Studies with proteinase inhibitors demonstrated that a serine proteinase is necessary for angiostatin generation. These data indicate that bioactive angiostatin can be generated directly by human prostate cancer cells and that serine proteinase activity is necessary for angiostatin generation.


Subject(s)
Neovascularization, Physiologic/drug effects , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Plasminogen/metabolism , Prostatic Neoplasms/metabolism , Serine Endopeptidases/physiology , Angiostatins , Humans , Male , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Tumor Cells, Cultured
10.
J Clin Invest ; 96(6): 2593-600, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675623

ABSTRACT

Expression of urokinase-type plasminogen activator (uPA) by malignant cells correlates with an aggressive phenotype, including increased invasiveness, tumor-associated angiogenesis, and metastases. Plasminogen activator inhibitor type 1 (PAI-1) is undetectable in cells of some aggressive malignancies, but present in the stroma of tumor-associated microvasculature. This analysis of an athymic mouse model of prostate carcinoma further defines the role of the uPA/PAI-1/plasmin system in primary growth and metastasis. A marked increase in PAI-1 expression was induced in clones of the aggressive human prostate carcinoma line, PC-3, by stable transfection. Primary PC-3 tumors, in mice, were significantly smaller when derived from PAI-1 expressing versus control cells. PAI-1 expression reduced the density of tumor-associated microvasculature by 22-38%. Microscopic metastases were quantitated using stable expression of the chromogenic label (beta-galactosidase) in control and PAI-1 expressing cells. PAI-1 expression resulted in a significant inhibition of lung metastases, and liver metastases. Expression of PAI-1 by malignant prostate cells resulted in a less aggressive phenotype, presumably by inhibition of uPA activity, suggesting pharmacologic or molecular inhibition of uPA activity as a potential therapeutic target.


Subject(s)
Liver Neoplasms/secondary , Lung Neoplasms/secondary , Neovascularization, Pathologic , Plasminogen Activator Inhibitor 1/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line , Clone Cells , Gene Expression , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Prostatic Neoplasms/blood supply , RNA, Messenger/biosynthesis , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , beta-Galactosidase/analysis , beta-Galactosidase/biosynthesis
11.
Neurosurgery ; 37(4): 723-30; discussion 730-2, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8559302

ABSTRACT

Basic fibroblast growth factor (bFGF) is a potent stimulator of angiogenesis, proliferation, and invasion in human gliomas. To test the hypothesis that bFGF is important in the development of the malignant phenotype of human gliomas, bFGF expression was prospectively modulated in primary human fetal astrocytes and in an established human glioma cell line. Fetal astrocytes were transfected with a vector expressing bFGF modified by the addition of a secretory signal peptide sequence. Two of these bFGF astrocyte clones examined in vitro demonstrated anchorage-independent growth, loss of contact inhibition, and decreased glial fibrillary acidic protein immunoreactivity, changes consistent with cellular transformation. To analyze the inhibition of bFGF expression, phosphorothioated bFGF antisense oligodeoxynucleotides were added to cultures of the U-87 human glioma cell line. The U-87 cell proliferation was inhibited to 70.6 +/- 0.4% of control at 10 mumol/L and to 53.2 +/- 5.6% of control at 20 mumol/L (P < 0.05). Both the 7.0- and 4.0-kilobase bFGF messenger ribonucleic acid transcripts were reduced after exposure to the antisense oligodeoxynucleotide, and cell-associated bFGF protein was reduced by 44%. The sense oligodeoxynucleotide, a negative control, failed to inhibit U-87 proliferation. These data support the concept that bFGF expression could be a key event in glial tumorigenesis that may be necessary for the sustained growth of human gliomas.


Subject(s)
Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Fibroblast Growth Factor 2/genetics , Transfection/genetics , Tumor Cells, Cultured/pathology , Astrocytes/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Transformed , Cell Transformation, Neoplastic/pathology , Fetus , Gene Expression Regulation, Neoplastic/physiology , Glial Fibrillary Acidic Protein/genetics , Glioma/genetics , Glioma/pathology , Humans , Transcription, Genetic/genetics
12.
Neurosurgery ; 36(4): 780-8, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7596510

ABSTRACT

Invasiveness is a critical event in the development of malignancy in brain tumors. A potential molecular mediator is basic fibroblast growth factor (bFGF). NIH-3T3 cells transfected with the bFGF gene fused with a signal peptide sequence (signal peptide bFGF) acquire an invasive phenotype as measured by in vitro assays of invasion including: 1) the formation of branching networks on Matrigel; 2) invasiveness in a chemoinvasion assay; 3) migration in a cell spreading assay; 4) detection of an Mr 92,000 gelatinase; and 5) local invasion into the surrounding neuropil after injection in the athymic mouse brain. By contrast, cells transfected with only the native bFGF gene (wild-type bFGF): 1) formed discrete cell clusters on Matrigel; 2) were less invasive and migratory in vitro; 3) released minimal Mr 92,000 collagenase; and 4) in vivo formed a pseudocapsule that separated the tumor cells from the neuropil. Quantitation of bFGF in the conditioned serum-free medium of the cell lines by enzyme-linked immunosorbent assay demonstrated that the signal peptide-bFGF cell clone secreted bFGF. These findings suggest a role for bFGF-mediated pathways and collagenase as molecular determinants of invasiveness in the brain.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Fibroblast Growth Factor 2/genetics , Neoplasm Invasiveness/genetics , Protein Sorting Signals/genetics , Recombinant Fusion Proteins/genetics , Transfection/genetics , 3T3 Cells , Animals , Cell Movement/genetics , Collagenases/genetics , Mice , Mice, Nude , Molecular Weight , Neoplasm Transplantation , Phenotype
13.
J Pharmacol Exp Ther ; 271(2): 1027-33, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7525934

ABSTRACT

Tumor growth is dependent upon angiogenesis. There is an intense search for pharmacological inhibitors of angiogenesis as a novel approach to treat angiogenic diseases, e.g., arthritis, diabetic retinopathy or cancer. A series of compounds, originally studied as potential protein kinase C inhibitors, included the diaminoanthraquinone NSC 639366 (1-[[3-(diethylamino)-2-hydroxypropyl]amino]-4-[(2,3- epoxypropyl)amino]-9,10-anthracenedione fumaric acid salt) (SPC-100097), was found to reversibly inhibit bovine endothelial cell growth with an IC50 that ranged between 1 and 4 nM. NSC 639366 reversibly inhibited endothelial cell migration, particularly endothelial cells stimulated by the potent angiogenic molecule, basic fibroblast growth factor. The activity of secreted urokinase-type plasminogen activator and active interstitial collagenase, but not gelatinase, was inhibited by NSC 639366. In vivo, angiogenesis was significantly inhibited by NSC 639366 by using the chick chorioallantoic membrane or the rat corneal bioassay. Two analogs of NSC 639366 did not inhibit endothelial cell growth. These experiments introduce a novel compound that could be clinically useful against angiogenic diseases and encourage further development of compounds that inhibit the plasminogen-plasmin system known to be a key regulator of angiogenesis.


Subject(s)
Anthraquinones/pharmacology , Neovascularization, Pathologic/prevention & control , Animals , Cattle , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Male , Metalloendopeptidases/metabolism , Plasminogen Activators/metabolism , Rats , Rats, Wistar
14.
Cancer Res ; 54(10): 2654-60, 1994 May 15.
Article in English | MEDLINE | ID: mdl-7513254

ABSTRACT

Suramin, an anticancer agent in current clinical trials, is a prototype of a pharmacological antagonist of growth factors, including basic fibroblast growth factor (bFGF). Suramin inhibited angiogenesis in the chick chorioallantoic membrane assay in a dose-dependent fashion. Suramin, 200 mg/kg i.v., inhibited rat corneal angiogenesis induced by bFGF-impregnated polymers; addition of heparin stimulated angiogenesis and counteracted the inhibition of suramin. The half-maximal inhibitory concentration (IC50) of suramin was determined for key cellular mechanisms that regulate angiogenesis: (a) low and high affinity cellular binding of bFGF to bovine capillary endothelial (BCE) cells with IC50s, respectively, of 24.3 and 71.5 micrograms/ml; (b) spontaneous migration of bovine pulmonary artery endothelial and normal AG 7680 fetal bovine aortic endothelial cells; bFGF-stimulated migration of BCE and transformed GM 7373 fetal bovine aortic endothelial cells with IC50s of 200-320 micrograms/ml; (c) proliferation of bovine pulmonary artery endothelial cells at > 100 micrograms/ml and of BCE cells at > 250 micrograms/ml; and (d) urokinase-type plasminogen activator activity of GM 7373 endothelial cells stimulated by bFGF with an IC50 of 211 micrograms/ml and of BCE cells stimulated by bFGF at > 100 micrograms/ml, but not plasminogen activator activity induced by phorbol 12-myristate 13-acetate. Suramin inhibited multiple control points of angiogenesis, including those stimulated by bFGF. Because tumor growth is angiogenesis dependent, the clinical efficacy of suramin may relate, in part, to angiosuppression.


Subject(s)
Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/metabolism , Neovascularization, Pathologic/prevention & control , Suramin/pharmacology , Urokinase-Type Plasminogen Activator/biosynthesis , Animals , Cell Division/drug effects , Cell Movement/drug effects , Chick Embryo , Cornea/blood supply , Drug Screening Assays, Antitumor , Endothelium, Vascular/pathology , Enzyme Induction/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Male , Rats , Rats, Wistar
15.
J Neurooncol ; 21(3): 189-201, 1994.
Article in English | MEDLINE | ID: mdl-7699415

ABSTRACT

Suramin is a novel anticancer agent that blocks the binding of growth factors, including basic fibroblast growth factor (bFGF), to their receptors. Prior studies showed human and experimental gliomas to upregulate and respond to autocrine stimulation by bFGF, the antiproliferative effects of suramin were therefore studied on glioma cell turnover in vitro and in the brain. Suramin inhibited the growth of rat (C6, 9L) and human (U-118, U-138, A-172, T98G) glioma cell lines in a dose-dependent manner. Suramin significantly reduced the bromodeoxyuridine (BUdR) labeling index of cultured glioma cells at 250 micrograms/ml, P < 0.0001. DNA flow cytometry revealed a significant decrease in the percentage of suramin-treated glioma cells in S-phase, P < 0.01. Using intracerebral rat C6 glioma model in vivo, suramin, 10-60 mg/kg, i.p., produced a dose-dependent reduction of BUdR labeling in both the glioma and endothelial cell subpopulations. Suramin, 200 mg/kg i.v., however, led to intratumoral hemorrhages that reduced survival. Electron microscopy revealed membranous inclusion bodies in the cytoplasm of C6 glioma and endothelial cells, an indication of excess glycosaminoglycans. Moreover, 46% of endothelial cells within the C6 glioma tumor treated with suramin, 60 mg/kg, i.p., developed membrane blebs. Suramin, in clinically relevant doses, significantly inhibits glioma cell growth and cytokinetics. The risk of intratumoral hemorrhage, possibly related to injury of endothelial cells or the accumulation of anticoagulant glycosaminoglycans, constitutes a major side effect and caution should be exercised in consideration of clinical application for intracerebral tumors.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioma/drug therapy , Suramin/pharmacology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flow Cytometry , Glioblastoma/pathology , Glioma/blood supply , Glioma/pathology , Humans , Male , Microscopy, Electron , Rats , Rats, Inbred WF , S Phase , Tumor Cells, Cultured/drug effects
16.
Cancer ; 70(11): 2673-80, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1423198

ABSTRACT

BACKGROUND: Microvascular proliferation, a prominent feature of tumors of the central nervous system, is a prime target for anti-cancer therapy. METHODS: Because basic fibroblast growth factor (bFGF) plays a key role in the regulation of angiogenesis, surgical specimens from 52 human brain tumors were examined by immunocytochemical studies with a murine monoclonal antibody to bFGF. Sections from these tumors also were incubated with Ki-67 monoclonal antibody to measure the growth fraction. RESULTS: Immunostaining for bFGF was observed in 45 of 52 (87%) neoplasms, reacting with 97% of the malignant brain tumors and 67% of benign tumors (P < 0.01). The nonreactive tumors were a medulloblastoma and 7 of 21 (33%) benign, noninvasive, slow-growing neoplasms (1 acoustic schwannoma, 3 meningiomas, 2 pituitary adenomas, and 1 cholesteatoma). The indices of proliferation (Ki-67 labeling) were lower for the 21 benign tumors (1.2 +/- 1.1%) than the 31 malignant tumors (10.3 +/- 10.5%; P < 0.001). The bFGF was immunolocalized in the tumor cell nuclei in 23 of 52 tumors (44%) and in the cytoplasm of 8 of 52 (15%) tumors. Immunostaining to bFGF was prominent in the microvascular endothelial compartment in 84% of the malignant tumors and only 52% of benign tumors (P < 0.01). Immunostaining was not present after preabsorption of the antibody with pure human recombinant bFGF. CONCLUSIONS: The presence of bFGF predominantly within the tumor microvasculature indicates a cellular depot for this potent growth factor that mediates angiogenesis and tumorigenesis. These data support a role for bFGF in the transition from the benign to the malignant phenotype.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/chemistry , Fibroblast Growth Factor 2/analysis , Microcirculation/chemistry , Blotting, Western , Cell Nucleus/chemistry , Frozen Sections , Humans , Immunohistochemistry , Ki-67 Antigen , Neoplasm Proteins/analysis , Nuclear Proteins/analysis , Tumor Cells, Cultured
17.
Cancer ; 68(4): 786-92, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1855178

ABSTRACT

Surgical specimens of 35 human brain tumors were examined with a novel monoclonal antibody, S-44, immunoreactive to statin, a nuclear protein specifically expressed in quiescent (noncycling) G0-phase cells. Benign tumors typically were statin positive with labeling indices (LI) between 22% and 96%: acoustic schwannomas (n = 3, mean = 29.9 +/- 19.4%); meningiomas (n = 4, mean = 59.0 +/- 15.1%); pituitary adenomas (n = 3, mean = 79.9 +/- 28.2%), and an epidermoid cyst (41.0%). By contrast, the statin LI of 18 of 24 (75%) malignant brain tumors was less than or equal to 2%: medulloblastomas (n = 7, mean = 0.3 +/- 0.2%); anaplastic astrocytomas (n = 3, mean = 1.6 +/- 2.7%); glioblastomas (n = 10, mean = 10.3 +/- 14.4%); metastatic carcinomas (n = 3, mean = 3.0 +/- 4.6); and a germinoma (0.2%). The vascular endothelium among diverse tumors typically was statin positive. All 21 tumors with a statin LI less than 10% were malignant, and all nine tumors with a statin LI greater than 40% were benign. The statin LI of benign tumors (n = 11, mean = 55.1 +/- 26.7%) was significantly higher than that of the malignant tumors (n = 24, mean = 5.2 +/- 10.5%, P less than 0.001). The absence of statin expression is a new way to determine the malignancy of human brain tumors. The statin LI may be useful to guide the prognosis and treatment of individual patients. The mechanisms that control statin expression are important in therapy seeking to shift the proliferating, cycling cells to the quiescent, G0 compartment.


Subject(s)
Brain Neoplasms/pathology , Proteins/analysis , Adolescent , Adult , Aged , Antibodies, Monoclonal , Biomarkers , Brain Neoplasms/chemistry , Cell Cycle Proteins , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Interphase , Male , Middle Aged , Peptide Elongation Factor 1 , Prospective Studies , Retrospective Studies
18.
Am J Pathol ; 137(5): 1121-42, 1990 Nov.
Article in English | MEDLINE | ID: mdl-1700617

ABSTRACT

Microvascular proliferation, a hallmark of malignant brain tumors, represents an attractive target of anticancer research, especially because of the quiescent nonproliferative endothelium of the normal brain. Cerebral neoplasms sequester copper, a trace metal that modulates angiogenesis. Using a rabbit brain tumor model, normocupremic animals developed large vascularized VX2 carcinomas. By contrast, small, circumscribed, relatively avascular tumors were found in the brains of rabbits copper-depleted by diet and penicillamine treatment (CDPT). The CDPT rabbits showed a significant decrease in serum copper, copper staining of tumor cell nuclei, microvascular density, the tumor volume, endothelial cell turnover, and an increase in the vascular permeability (breakdown of the blood-brain barrier), as well as peritumoral brain edema. In non-tumor-bearing animals, CDPT did not alter the vascular permeability or the brain water content. CDPT also inhibited the intracerebral growth of the 9L gliosarcoma in F-344 rats, with a similar increase of the peritumoral vascular permeability and the brain water content. CDPT failed to inhibit tumor growth and the vascularization of the VX2 carcinoma in the thigh muscle or the metastases to the lung, findings that may reflect regional differences in the responsiveness of the endothelium, the distribution of copper, or the activity of cuproenzymes. Metabolic and pharmacologic withdrawal of copper suppresses intracerebral tumor angiogenesis; angiosuppression is a novel biologic response modifier for the in situ control of tumor growth in the brain.


Subject(s)
Brain Neoplasms/pathology , Copper/deficiency , Neovascularization, Pathologic/pathology , Penicillamine/therapeutic use , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/therapy , Bromodeoxyuridine , Capillary Permeability , Cell Division , Copper/blood , Diet , Fibronectins/analysis , Glioma/blood , Glioma/pathology , Glioma/therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Rabbits , Rats
SELECTION OF CITATIONS
SEARCH DETAIL