Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 326
Filter
1.
J Immunol Methods ; 342(1-2): 78-81, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19109967

ABSTRACT

Many of the current in vivo methods to evaluate angiogenesis are poorly quantifiable. Recently, the Matrigel plug assay has become the method of choice in many studies involving in vivo testing for angiogenesis. When known angiogenic factors are mixed with Matrigel and injected subcutaneously into mice, endothelial cells migrate into the gel plug. These endothelial cells form vessel-like structures, a process that mimics the formation of capillary networks. Here, we present a modification of the traditional Matrigel assay with improved method to quantify the amount of endothelial cells that incorporate into the plug. The removed plugs were subjected to a mild protease treatment, yielding intact cells. The liberated cells were then stained using an endothelial cell-specific markers, and counted by flow cytometry. This novel combination of FACS analysis with the traditional Matrigel assay improves the ability to quantify in vivo angiogenesis, and for the first time enables to determine the number of migrating and proliferating endothelial cells which reflects the angiogenesis rate.


Subject(s)
Collagen/pharmacology , Endothelial Cells/drug effects , Laminin/pharmacology , Neovascularization, Physiologic/drug effects , Proteoglycans/pharmacology , Angiogenesis Inducing Agents/pharmacology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Drug Combinations , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry/methods , Humans , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/pharmacology
2.
Cancer Gene Ther ; 13(6): 619-27, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16456550

ABSTRACT

Developing continuous systemic delivery of endostatin has been a goal of many laboratories. We have employed a method of gene therapy utilizing different viral constructs. Here, we report that a new serotype of adeno-associated viruses, which incorporates canine endostatin, provides dose-dependent transgene expression in the circulation after intramuscular injection in mice. Elevated levels of endostatin remained stable in the circulation for at least 4 months. In vitro assays determined that the protein expressed was biologically active. Antitumor activities of the above construct demonstrated a U-shape curve, where the maximum activity was observed within a certain critical concentration range. These data suggest that an optimum dose range may be required to achieve therapeutic efficacy in large animal models.


Subject(s)
Antineoplastic Agents/therapeutic use , Dependovirus/genetics , Endostatins/therapeutic use , Genetic Therapy/methods , Neoplasms/drug therapy , Amino Acid Sequence , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Endostatins/administration & dosage , Endostatins/genetics , Genetic Vectors , Humans , Injections, Intramuscular , Male , Mice , Mice, SCID , Molecular Sequence Data , Pancreatic Neoplasms/drug therapy , Sequence Alignment
3.
Article in English | MEDLINE | ID: mdl-16869776

ABSTRACT

Does an activated oncogene that initiates tumor growth need to remain activated to maintain the cancer phenotype? This question has been answered affirmatively by experiments in which doxycycline-regulated oncogene activation induces growth of large tumors that regress completely upon oncogene inactivation--a phenomenon called oncogene addiction. We assemble here the evidence that oncogene addiction is angiogenesis-dependent. Although activated oncogenes increase tumor cell proliferation and decrease their apoptosis, these activities are not sufficient to expand tumor mass beyond a microscopic size. Oncogenes must also induce tumor angiogenesis for expansion of tumor mass. We propose experiments to validate the "endothelial centric" hypothesis of oncogene addiction.


Subject(s)
Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Oncogenes , Animals , Apoptosis , Cell Proliferation , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Humans , Mice , Mice, SCID , Mice, Transgenic , Models, Biological , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Oncogenes/drug effects , Thrombospondin 1/metabolism
5.
Biochem Biophys Res Commun ; 292(4): 886-91, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11944897

ABSTRACT

Angiostatin is a potent inhibitor of angiogenesis generated in cancer-bearing hosts by tumor-derived proteases. Because the naturally occurring bone and prostate cancers of pet dogs provide unique model systems to study factors that regulate cancer progression and tumor dormancy, we investigated the capacity of these tumors to generate angiostatin. We determined that angiostatin fragments are present in urine of dogs with bone cancer. The identity of these fragments was confirmed by comparison of the experimentally determined protein sequence to that of a clone of canine angiostatin. Importantly, these fragments were absent in urine collected from the same dogs after complete surgical removal of the primary tumor. We also demonstrate that canine prostate cancer cells are capable of processing plasminogen to angiostatin in vitro. These findings provide rationale for using spontaneous canine tumor models to isolate endogenous angiogenesis inhibitors and to investigate their therapeutic use against cancer.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/metabolism , Osteosarcoma/veterinary , Peptide Fragments/metabolism , Plasminogen/metabolism , Prostatic Neoplasms/veterinary , Angiostatins , Animals , Antibody Specificity , Bone Neoplasms/metabolism , Cattle , Cell Division/drug effects , Disease Progression , Dogs , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Male , Molecular Sequence Data , Osteosarcoma/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/urine , Plasminogen/chemistry , Plasminogen/genetics , Plasminogen/urine , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sequence Homology, Amino Acid , Tumor Cells, Cultured
6.
Semin Oncol ; 28(6): 536-42, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11740806

ABSTRACT

There is accumulating evidence that leukemias and other hematologic diseases are angiogenesis-dependent. Therefore, angiogenesis inhibitors may be useful adjuncts to conventional therapies in the treatment of these diseases. When all conventional therapy has failed, an angiogenesis inhibitor may be successfully used alone, as has been demonstrated in the treatment of multiple myeloma by thalidomide. The experimental and clinical evidence that the efficacy of thalidomide against multiple myeloma is mediated in part by its antiangiogenic activity is presented here. In current clinical trials throughout the United States, angiogenesis inhibitors are being used against cancer in combination with conventional chemotherapy or radiotherapy. Angiogenesis inhibitors are also being used in combination with each other. This practice may increase as different angiogenesis inhibitors become more widely available.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Multiple Myeloma/blood supply , Multiple Myeloma/drug therapy , Thalidomide/therapeutic use , Animals , Humans , Leukemia/pathology , Neovascularization, Pathologic , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
Cancer Res ; 61(19): 7298-304, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585769

ABSTRACT

A primary inoculum of human pancreatic cancer cells (BxPC-3) has the ability to inhibit the growth of a secondary tumor in an in vivo animal model. Such ability suggests that the primary tumor is producing inhibitors that act at the site of the secondary tumor. Accordingly we attempted to discover which inhibitors are produced by pancreatic cancer cells. We determined that pancreatic cancer cells process angiostatin isoforms from plasminogen. Additionally, we isolated and characterized an uncleaved "latent" antiangiogenic antithrombin (aaAT) molecule processed from systemically available AT by pancreatic cancer cells as well as a cleaved form of aaAT processed from systemically available AT by pancreatic cancer cells. Human AT, cleaved with human neutrophil elastase, inhibits angiogenesis in the chorioallantoic membrane assay. This human aaAT molecule is able to inhibit the growth of pancreatic tumors in immune-compromised mice. Our work represents the first demonstration of multiple angiogenesis inhibitors from a single tumor and suggests that antiangiogenic therapies may provide an avenue for future treatment of pancreatic cancer.


Subject(s)
Adenocarcinoma/metabolism , Angiogenesis Inhibitors/biosynthesis , Antithrombins/biosynthesis , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/metabolism , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Adenocarcinoma/blood , Adenocarcinoma/blood supply , Angiogenesis Inhibitors/isolation & purification , Angiogenesis Inhibitors/pharmacology , Angiostatins , Animals , Antithrombins/isolation & purification , Antithrombins/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Cell Division/physiology , Chick Embryo , Culture Media, Conditioned , Endothelium, Vascular/cytology , Fibrosarcoma/blood supply , Fibrosarcoma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neovascularization, Physiologic/drug effects , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/blood supply , Plasminogen/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Res ; 61(20): 7669-74, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11606410

ABSTRACT

In the first Phase I clinical trials of endostatin as an antiangiogenic therapy for cancer, the protein was administered as an i.v. bolus for approximately 20-30 min each day. This protocol was based on experimental studies in which animals were treated by s.c. bolus once a day. However, it was not clear in the previous studies whether this schedule could be maximized further. Therefore, we developed experimental models involving continuous administration of endostatin to determine the potency and efficacy of this approach. Endostatin was administered to tumor-bearing mice either s.c. or i.p. in single bolus doses. The efficacy of these regimens was compared with endostatin administered continuously via an i.p. implanted mini-osmotic pump. Our results show that endostatin remains stable and active in mini-osmotic pumps for at least 7 days. We show that endostatin injected i.p. is rapidly cleared within 2 h, whereas endostatin administered continuously via mini-osmotic pump maintains systemic concentrations of 200-300 ng/ml for the duration of administration. Furthermore, continuous i.p. administration of endostatin results in more effective tumor suppression at significantly reduced doses (5-fold), compared with bolus administration. Additional experiments using a human pancreatic cancer model in severe combined immunodeficient mice showed that there was a significant decrease in the microvessel density between the treatment groups and the control group. These data show that continuous administration of human endostatin results in sustained systemic concentrations of the protein leading to: (a) increased efficacy manifested as increased tumor regression; and (b) an 8-10-fold decrease in the dose required to achieve the same antitumor effect as the single daily bolus administration of endostatin. On the basis of this approach, an additional clinical trial has been designed and initiated and is under way in two countries.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Collagen/administration & dosage , Peptide Fragments/administration & dosage , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Collagen/pharmacokinetics , Drug Stability , Endostatins , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Humans , Infusion Pumps, Implantable , Infusions, Parenteral , Injections, Intraperitoneal , Male , Mice , Mice, Inbred CBA , Mice, SCID , Neovascularization, Pathologic/drug therapy , Osmotic Pressure , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Peptide Fragments/pharmacokinetics , Xenograft Model Antitumor Assays
11.
Thromb Haemost ; 86(1): 23-33, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11487011

ABSTRACT

Angiogenesis research is being translated to the clinic. Certain guidelines may facilitate this effort. Recruitment of endothelial cells by a tumor is an early event in angiogenesis, a process regulated at genetic and epigenetic levels. The microvascular endothelial cell has become an important second target in cancer therapy. Angiogenesis inhibitors are either "direct" or "indirect" and their optimal dosing depends on a different logic than conventional chemotherapy. Conversely, antiangiogenic scheduling of chemotherapy can by-pass drug resistance. Like all solid tumors, hematologic malignancies are angiogenesis-dependent. Further, angiogenesis is modulated by proteins and cells from the hematopoietic and hemostatic systems. Clinical testing of angiogenesis inhibitors has accentuated the need for surrogate markers of tumor angiogenesis activity. Microvessel density, so valuable as a prognostic indicator of metastatic risk, cannot determine efficacy of an angiogenesis inhibitor. In the future, angiogenesis inhibitors may be added to chemotherapy or to radiotherapy, or to other modalities. Also, combinations of angiogenesis inhibitors may be administered together.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Hemostatics/pharmacology , Humans , Leukemia , Neoplasms/drug therapy , Neovascularization, Pathologic/etiology , Practice Guidelines as Topic
13.
J Natl Cancer Inst ; 93(14): 1075-81, 2001 Jul 18.
Article in English | MEDLINE | ID: mdl-11459868

ABSTRACT

BACKGROUND: Tumor cells are known to be heterogeneous with respect to their metastatic activity, proliferation rate, and activity of several enzymes. However, little is known about the heterogeneity of tumor angiogenic activity. We investigated whether heterogeneity of angiogenic activity could be responsible for the well-known observation of "no take" of human tumors transplanted into immunodeficient mice. METHODS: Severe combined immunodeficient (SCID) mice were xenotransplanted subcutaneously with tumor tissue (n = 55) or cell suspension of a human liposarcoma cell line (SW-872) or subclones (n = 28), with varying cell proliferation rates. Xenograft tumor growth was recorded for up to 6 months. Tumor tissues were then removed and analyzed for tumor cell apoptosis, microvessel density, and cell proliferation. All statistical tests were two-sided. RESULTS: Pieces of tumor derived from the parental cell line or its clones gave rise to three kinds of tumors: 1) highly angiogenic and fast-growing (aggressive) tumors, 2) weakly angiogenic and slow-growing tumors, and 3) nonangiogenic and stable tumors. Most tumors retained the original phenotype of their parental tumor. Tumor volume correlated positively with microvessel density (Spearman correlation coefficient [r] =.89; P< or =.0001) and inversely with tumor cell apoptosis (Spearman r = -.68; P =.002). Tumor volume was less strongly but still positively correlated with tumor cell proliferation in vivo (Spearman r =.55; P =.02). CONCLUSIONS: Human liposarcoma cells appear to be heterogeneous in their angiogenic activity. When tumor cells with little or no angiogenic activity are transplanted into SCID mice, a microscopic, dormant tumor results that may not grow further. Because such tiny tumors are neither grossly visible nor palpable, they have previously been called "no take." The finding that an angiogenic tumor can contain subpopulations of tumor cells with little or no angiogenic activity may provide a novel mechanism for dormant micrometastases, late recurrence, and changes in rate of tumor progression.


Subject(s)
Disease Models, Animal , Liposarcoma/blood supply , Neoplasm Transplantation , Neovascularization, Pathologic , Animals , Apoptosis , Cell Division , Humans , Immunohistochemistry , Mice , Mice, SCID , Phenotype , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
16.
Proc Natl Acad Sci U S A ; 98(8): 4605-10, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11274374

ABSTRACT

Although the systemic administration of a number of different gene products has been shown to result in the inhibition of angiogenesis and tumor growth in different animal tumor models, the relative potency of those gene products has not been studied rigorously. To address this issue, recombinant adenoviruses encoding angiostatin, endostatin, and the ligand-binding ectodomains of the vascular endothelial growth factor receptors Flk1, Flt1, and neuropilin were generated and used to systemically deliver the different gene products in several different preexisting murine tumor models. Single i.v. injections of viruses encoding soluble forms of Flk1 or Flt1 resulted in approximately 80% inhibition of preexisting tumor growth in murine models involving both murine (Lewis lung carcinoma, T241 fibrosarcoma) and human (BxPC3 pancreatic carcinoma) tumors. In contrast, adenoviruses encoding angiostatin, endostatin, or neuropilin were significantly less effective. A strong correlation was observed between the effects of the different viruses on tumor growth and the activity of the viruses in the inhibition of corneal micropocket angiogenesis. These data underscore the need for comparative analyses of different therapeutic approaches that target tumor angiogenesis and provide a rationale for the selection of specific antiangiogenic gene products as lead candidates for use in gene therapy approaches aimed at the treatment of malignant and ocular disorders.


Subject(s)
Neovascularization, Pathologic , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Transfection , Adenoviridae/genetics , Amino Acid Sequence , Animals , Cell Division/genetics , Evaluation Studies as Topic , Humans , Mice , Molecular Sequence Data , Neoplasms/blood supply , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-1
17.
Cancer Res ; 61(5): 2207-11, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280788

ABSTRACT

The surgical removal of a primary tumor can result in the rapid growth of metastases. The production of angiogenesis inhibitors by the primary tumor is one mechanism for the inhibition of metastatic tumor growth. The effect of curative radiotherapy to a primary tumor known to make an inhibitor of angiogenesis and the effects on distant metastases has not been studied. We here show that the eradication of a primary Lewis lung carcinoma (LLC-LM), which is known to generate angiostatin, is followed by the rapid growth of metastases that kill the animal within 18 days after the completion of radiation therapy. The right thighs of C57BL/6 mice (n = 25) were injected s.c. with 1 x 10(6) LLC-LM cells. Animals were randomized to one of five groups: no irradiation, 40 Gy in one fraction, 30 Gy in one fraction, 40 Gy in two 20 Gy fractions, or 50 Gy in five 10 Gy fractions. Tumors were clinically eradicated in each treatment group. All of the surviving animals became dyspneic and were killed within 14-18 days after the completion of radiation therapy. Examination of their lungs revealed >46 (range, 46-62) surface metastases in the treated animals compared with 5 (range, 2-8) in the untreated animals. The lung weights had increased from 0.2 g (range, 0.19-0.22 g) in the controls to 0.58 g (range 0.44-0.84) in the experimental animals. The most effective dose regimen was 10 Gy per fraction for five fractions, and serial experiments were conducted with this fractionation scheme. Complete response of the primary tumor was seen in 25 of 35 (71%) mice. The average weight of the lungs in the nonirradiated animals was 0.22 g (range, 0.19-0.24 g) and in the irradiated animals was 0.66 g (range, 0.61-0.70 g). The average number of surface metastases increased from five per lung (range, 2-13) in the control animals to 53 per lung (range, 46-62) in the irradiated animals. Both differences were statistically significant with P < 0.001. If the nontumor-bearing leg was irradiated or the animals were sham-irradiated, no difference in the number of surface metastases or lung weights was observed between the control group and the treated group. Urinary levels of matrix metalloproteinase 2, the enzyme responsible for angiostatin processing in this tumor model, were measured and correlated with the viability and size of the primary tumor. Administration of recombinant angiostatin prevented the growth of the metastases after the treatment of the primary tumor. In this model, the use of radiation to eradicate a primary LLC-LM tumor results in the growth of previously dormant lung metastases and suggests that combining angiogenesis inhibitors with radiation therapy may control distant metastases.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Lewis Lung/secondary , Fibrosarcoma/radiotherapy , Fibrosarcoma/secondary , Angiogenesis Inhibitors/pharmacology , Angiostatins , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Cell Division/radiation effects , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Male , Matrix Metalloproteinase 2/urine , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Peptide Fragments/biosynthesis , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Plasminogen/biosynthesis , Plasminogen/pharmacology , Plasminogen/physiology , Radiotherapy/adverse effects
18.
J Cell Biol ; 152(6): 1233-46, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11257123

ABSTRACT

Collagen XVIII (c18) is a triple helical endothelial/epithelial basement membrane protein whose noncollagenous (NC)1 region trimerizes a COOH-terminal endostatin (ES) domain conserved in vertebrates, Caenorhabditis elegans and Drosophila. Here, the c18 NC1 domain functioned as a motility-inducing factor regulating the extracellular matrix (ECM)-dependent morphogenesis of endothelial and other cell types. This motogenic activity required ES domain oligomerization, was dependent on rac, cdc42, and mitogen-activated protein kinase, and exhibited functional distinction from the archetypal motogenic scatter factors hepatocyte growth factor and macrophage stimulatory protein. The motility-inducing and mitogen-activated protein kinase-stimulating activities of c18 NC1 were blocked by its physiologic cleavage product ES monomer, consistent with a proteolysis-dependent negative feedback mechanism. These data indicate that the collagen XVIII NC1 region encodes a motogen strictly requiring ES domain oligomerization and suggest a previously unsuspected mechanism for ECM regulation of motility and morphogenesis.


Subject(s)
Bacterial Proteins , Cell Movement/physiology , Collagen/metabolism , Endothelium, Vascular/cytology , Extracellular Matrix/physiology , Peptide Fragments/metabolism , Protein Structure, Tertiary , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Bacterial Toxins/pharmacology , Blotting, Western , Cell Movement/drug effects , Cells, Cultured , Collagen/chemistry , Collagen/genetics , Collagen Type XVIII , Cytotoxins/pharmacology , Dimerization , Endostatins , Endothelium, Vascular/drug effects , Endothelium, Vascular/growth & development , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
19.
J Natl Cancer Inst ; 93(5): 382-7, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11238700

ABSTRACT

BACKGROUND: Angiogenesis is essential for tumor growth and progression. Therefore, inhibition of angiogenesis is being studied as a new anticancer therapy. Because cytotoxic chemotherapy is more effective on rapidly growing tumors than on slowly growing tumors, it has been assumed that antiangiogenic therapy will also be effective only on rapidly growing, highly vascularized tumors. We compared the effects of two angiogenesis inhibitors, TNP-470 and angiostatin, on slowly growing, poorly vascularized and rapidly growing, highly vascularized human tumors in mice. METHODS: Slowly growing (RT-4) and rapidly growing (MGH-U1) human bladder carcinoma cell lines were grown in severe combined immunodeficiency mice. Established tumors were treated with one of the two angiogenesis inhibitors. Tumor volumes, vascularity, and proliferation indices were determined. The in vitro effects of TNP-470 and of angiostatin on the proliferation of RT-4 and MGH-U1 cells were also investigated. All statistical tests were two-sided. RESULTS: RT-4 and MGH-U1 tumor growth was statistically significantly inhibited by both angiogenesis inhibitors (P<.001). Both inhibitors decreased the blood vessel density in both tumor types but did not alter the in vivo proliferation indices of the tumors. TNP-470, but not angiostatin, marginally decreased the in vitro proliferation of MGH-U1 cells. CONCLUSION: Slowly growing, poorly vascularized tumors in animal models respond as well as rapidly growing, highly vascularized tumors to therapy with the angiogenesis inhibitors TNP-470 and angiostatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Sesquiterpenes/pharmacology , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/drug therapy , Angiostatins , Animals , Carcinoma/blood supply , Carcinoma/drug therapy , Cyclohexanes , Humans , Immunohistochemistry , Mice , Mice, SCID , O-(Chloroacetylcarbamoyl)fumagillol
20.
Dev Biol ; 230(2): 139-50, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11161568

ABSTRACT

The transmembrane ligand ephrinB2 and its receptor tyrosine kinase EphB4 are molecular markers of embryonic arterial and venous endothelial cells, respectively, and are essential for angiogenesis. Here we show that expression of ephrinB2 persists in adult arteries where it extends into some of the smallest diameter microvessels, challenging the classical view that capillaries have neither arterial nor venous identity. EphrinB2 also identifies arterial microvessels in several settings of adult neovascularization, including tumor angiogenesis, contravening the dogma that tumor vessels arise exclusively from postcapillary venules. Unexpectedly, expression of ephrinB2 also defines a stable genetic difference between arterial and venous vascular smooth muscle cells. These observations argue for revisions of classical concepts of capillary identity and the topography of neovascularization. They also imply that ephrinB2 may be functionally important in neovascularization and in arterial smooth muscle, as well as in embryonic angiogenesis.


Subject(s)
Arteries/cytology , Arterioles/pathology , Endothelium, Vascular/pathology , Lung Neoplasms/blood supply , Melanoma, Experimental/blood supply , Membrane Proteins/genetics , Microcirculation/pathology , Muscle, Smooth, Vascular/pathology , Neovascularization, Pathologic/pathology , Veins/cytology , Venules/pathology , Animals , Arteries/metabolism , Arteries/pathology , Arterioles/metabolism , Biomarkers , Endothelium, Vascular/metabolism , Ephrin-B2 , Membrane Proteins/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Reference Values , Veins/metabolism , Veins/pathology , Venules/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...