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










Publication year range
1.
Sci Immunol ; 9(94): eadh2334, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669316

ABSTRACT

T cells are often absent from human cancer tissues during both spontaneously induced immunity and therapeutic immunotherapy, even in the presence of a functional T cell-recruiting chemokine system, suggesting the existence of T cell exclusion mechanisms that impair infiltration. Using a genome-wide in vitro screening platform, we identified a role for phospholipase A2 group 10 (PLA2G10) protein in T cell exclusion. PLA2G10 up-regulation is widespread in human cancers and is associated with poor T cell infiltration in tumor tissues. PLA2G10 overexpression in immunogenic mouse tumors excluded T cells from infiltration, resulting in resistance to anti-PD-1 immunotherapy. PLA2G10 can hydrolyze phospholipids into small lipid metabolites, thus inhibiting chemokine-mediated T cell mobility. Ablation of PLA2G10's enzymatic activity enhanced T cell infiltration and sensitized PLA2G10-overexpressing tumors to immunotherapies. Our study implicates a role for PLA2G10 in T cell exclusion from tumors and suggests a potential target for cancer immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes , Up-Regulation , Animals , Female , Humans , Mice , Cell Line, Tumor , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Neoplasms/immunology , Phospholipases A/immunology , Phospholipases A/genetics , Phospholipases A2/immunology , T-Lymphocytes/immunology , Up-Regulation/immunology
2.
Mol Cell Biol ; 29(17): 4663-78, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564416

ABSTRACT

Uncontrolled fibroblast growth factor (FGF) signaling can lead to human diseases, necessitating multiple layers of self-regulatory control mechanisms to keep its activity in check. Herein, we demonstrate that FGF9 and FGF20 ligands undergo a reversible homodimerization, occluding their key receptor binding sites. To test the role of dimerization in ligand autoinhibition, we introduced structure-based mutations into the dimer interfaces of FGF9 and FGF20. The mutations weakened the ability of the ligands to dimerize, effectively increasing the concentrations of monomeric ligands capable of binding and activating their cognate FGF receptor in vitro and in living cells. Interestingly, the monomeric ligands exhibit reduced heparin binding, resulting in their increased radii of heparan sulfate-dependent diffusion and biologic action, as evidenced by the wider dilation area of ex vivo lung cultures in response to implanted mutant FGF9-loaded beads. Hence, our data demonstrate that homodimerization autoregulates FGF9 and FGF20's receptor binding and concentration gradients in the extracellular matrix. Our study is the first to implicate ligand dimerization as an autoregulatory mechanism for growth factor bioactivity and sets the stage for engineering modified FGF9 subfamily ligands, with desired activity for use in both basic and translational research.


Subject(s)
Extracellular Matrix/metabolism , Fibroblast Growth Factor 9/chemistry , Fibroblast Growth Factor 9/metabolism , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Heparitin Sulfate/metabolism , Protein Structure, Quaternary , Amino Acid Sequence , Animals , Cells, Cultured , Crystallography, X-Ray , Diffusion , Dimerization , Extracellular Matrix/chemistry , Female , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factors/genetics , Humans , Ligands , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutation , Pregnancy , Protein Multimerization , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/physiology
3.
Int J Radiat Biol ; 84(5): 401-12, 2008 May.
Article in English | MEDLINE | ID: mdl-18464069

ABSTRACT

PURPOSE: Velafermin (recombinant human fibroblast growth factor-20, rhFGF-20) has been shown to reduce the severity and duration of mucositis in preclinical acute (single dose) radiation and chemotherapy/radiation models of oral mucositis. Our present study assessed the impact of velafermin on the severity and duration of oral mucositis that occurred as a consequence of fractionated radiation. EXPERIMENTAL DESIGN: Male Golden Syrian hamsters were exposed to eight doses of radiation (7.5 Gy/dose) to the cheek pouch on days 0, 1, 2, 3, 6, 7, 8 and 9 that resulted in severe mucositis. Velafermin (4 mg/kg intraperitoneally) was administered on days 3 and 9; days 2, 3, 8 and 9; days 3, 4, 9 and 10; or days 4, 5, 10 and 11. RESULTS: Although all velafermin-treated groups showed some reduction in the degree of mucositis relative to the vehicle control, the most significant reduction (p < 0.001) was observed in the groups treated on days 3 and 9 or on days 4, 5, 10 and 11. Further histological analysis of resected buccal mucosa revealed improvements in epithelial tissue degradation, connective tissue degradation and inflammation severity after velafermin treatment. Most notably, velafermin treatment reduced inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor (TNF) production possibly through nuclear factor-kappaB (NF-kappaB) mediation. The detection of increased NF-E2-related factor-2 (NRF-2) expression in the early onset stage of mucositis in the buccal mucosa suggested additional protective benefits from reactive oxygen species (ROS) generated as a consequence of fractionated radiation treatment. CONCLUSION: Thus, velafermin provided therapeutic benefit in a hamster model of oral mucositis induced by fractionated radiation therapy.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/pharmacology , Mouth Mucosa/pathology , Radiation Injuries/drug therapy , Stomatitis/drug therapy , Animals , Cheek , Cricetinae , Epithelium/radiation effects , Humans , Inflammation/radiotherapy , Interleukin-6/biosynthesis , Male , NF-E2-Related Factor 2/biosynthesis , NF-kappa B/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
4.
Mol Oncol ; 2(1): 81-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19383330

ABSTRACT

GPNMB is a melanoma-associated glycoprotein that is targeted by the CR011-vcMMAE antibody-drug conjugate (ADC). Previous studies have shown that CR011-vcMMAE induces the apoptosis of GPNMB-expressing tumor cells in vitro and tumor regression in xenograft models. This ADC is currently in clinical trials for melanoma. In the present investigation, a variety of compounds were examined for their ability to increase the expression of GPNMB by cancer cells. These experiments lead to the identification of three distinct groups of compounds that increased GPNMB, some of which were shown to enhance the sensitivity of melanoma cells to CR011-vcMMAE. These data indicate that it may be possible to increase the anticancer activity of CR011-vcMMAE through pharmacological enhancement of GPNMB expression for potential therapeutic benefit.


Subject(s)
Antibodies, Monoclonal/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation/drug effects , Glioma/drug therapy , Immunoconjugates/pharmacology , Melanoma/drug therapy , Membrane Glycoproteins/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Glioma/pathology , Humans , Immunoconjugates/therapeutic use , Melanoma/pathology
5.
Int J Cancer ; 122(6): 1400-10, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18027850

ABSTRACT

Histone deacetylase inhibitors (HDACi) represent a promising new class of anticancer agents. In the current investigation, we examined the activity of the HDACi belinostat in preclinical models of prostate cancer. In vitro proliferation assays demonstrated that belinostat potently inhibited the growth of prostate cancer cell lines (IC(50) < 1.0 microM) and was cytotoxic to these cells. Washout experiments indicated that exposure to belinostat for relatively short periods of time (<12 hr) induced suboptimal growth-inhibition and that cells exposed to 1.0 microM belinostat for 48 hr retained the capacity for regrowth following drug withdrawal, while cells exposed to 4.0 microM belinostat were irreversibly growth-inhibited. Cell cycle analyses demonstrated that belinostat induced G2/M arrest and increased the percentage of cells with subG1 DNA content, thus confirming the growth-inhibitory and cytotoxic effects of this compound. Normal prostate epithelial cells were generally less susceptible to the effects of belinostat than were prostate cancer cells. In an orthotopic prostate cancer tumor model, belinostat inhibited tumor growth by up to 43%. Moreover, metastatic lung lesions were present in 47% of vehicle-treated animals but in none of the animals administered belinostat. Consistent with its observed antimetastatic activity, belinostat inhibited the migration of prostate tumor cells and increased the production of tissue inhibitor of metalloproteinase-1 (TIMP-1) by these cells, the latter effect being replicated by siRNA knockdown of HDAC3. Belinostat also increased the expression of p21 and decreased the expression of potentially oncogenic proteins (mutant p53 and ERG). These results support the clinical evaluation of belinostat for the treatment of prostate cancer.


Subject(s)
Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Cell Division/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Sulfonamides
6.
Dev Dyn ; 236(12): 3459-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17969154

ABSTRACT

Heterogeneous epithelial populations comprising the thymic environment influence early and late stages of T-cell development. The processes that regulate the differentiation of thymic epithelium and that are responsible for this heterogeneity are not well understood, although mesenchymal/epithelial interactions are clearly involved. Here, we show that targeted expression by thymocytes of an fibroblast growth factor receptor-2IIIb (FGFR2IIIb) ligand, FGF10, profoundly alters the differentiation and function of thymic epithelium (TE). Reconstitution of irradiated lckFGF10 mice with normal bone marrow restores normal thymic organization and function, while wild-type mice reconstituted with lckFGF10 bone marrow recapitulates some of the thymic alterations seen in lckFGF10 mice. We also demonstrate that interference with FGFR2IIIb signaling in the thymus with a soluble FGFR2IIIb dominant-negative fusion protein leads to precocious reductions in thymic size and cellularity that resemble age-related thymic involution. These findings indicate that TE compartments are dynamically maintained and that FGF signals are involved in this process.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2/physiology , Thymus Gland/growth & development , Aging/metabolism , Aging/pathology , Animals , Base Sequence , DNA Primers/genetics , Epithelium/growth & development , Epithelium/metabolism , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/physiology , Gene Expression Regulation, Developmental , Gene Targeting , Humans , Keratins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/metabolism
7.
Cancer Chemother Pharmacol ; 60(3): 423-35, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17541593

ABSTRACT

PURPOSE: To investigate the pharmacological properties of the CR011-vcMMAE fully human antibody-drug conjugate (ADC), such as dose titrations, quantitation of the time (days) to complete regression, pharmacokinetics, and schedule dependency. Our prior study characterized a fully human antibody to GPNMB covalently linked to monomethylauristatin E, CR011-vcMMAE, and further demonstrated cell surface staining of melanoma lines susceptible to the immunoconjugate's cytotoxicity (Clin Cancer Res 2005; 12(4): 1373-1382). METHODS: The human SK-MEL-2 and SK-MEL-5 melanoma xenografts were used in athymic mice to assess anti-tumor efficacy. After s.c. implantation, tumors became established (60-100 mg), and treatment commenced by i.v. injection of the immunoconjugate or vinblastine or paclitaxel. Short-term anti-tumor effects (inhibition of tumor growth) and long-term effects (complete regression) were observed. RESULTS: CR011-vcMMAE induced regression of established human SK-MEL-2 and SK-MEL-5 xenografts at doses from 1.25 to 80 mg/kg treatment when administered intravenously every 4 days (4 treatments); strikingly, regressions were not associated with re-growth during the observation period (200 days). The disappearance rate of implants was dose dependent (minimum time, 18.5 days). Detectable serum CR011-vcMMAE >or=1 microg/mL (approximately 0.01 microM) was observed for >30 days post-dose; CR011-vcMMAE showed an elimination half-life of 10.3 days. A low volume of distribution suggested that CR011-vcMMAE was confined to blood and interstitial fluid. CR011-vcMMAE could be delivered by either a single bolus dose or by intermittent dosing (i.e., every 1, 2, 4, 8, or 16 days) with no discernible differences in the proportion of tumor-free survivors, indicating a lack of schedule dependency. The antibody-drug conjugate produced complete regressions, but the equivalent doses of free monomethylauristatin E or unconjugated antibody did not show anti-tumor effects. In addition, decreases in plasma tumor-derived human interleukin-8 coincided with tumor nodule disappearance. CONCLUSIONS: Short-term anti-tumor effects and long-term effects (complete regression) were observed with CR011-vcMMAE, but not with the reference agents. These results suggest that CR011-vcMMAE may provide therapeutic benefit in malignant melanoma.


Subject(s)
Immunotoxins/therapeutic use , Melanoma/drug therapy , Membrane Glycoproteins/drug effects , Adult , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Melanoma/pathology , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis/drug therapy , Transplantation, Heterologous
8.
Nephrol Dial Transplant ; 22(5): 1323-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17308324

ABSTRACT

BACKGROUND: Arresting or regressing kidney scarring is of major clinical relevance. Platelet-derived growth factor D (PDGF-D) is widely expressed in fibrotic kidneys. Administration of the PDGF-D neutralizing fully human monoclonal antibody CR002 in the acute phase of progressive anti-Thy 1.1 glomerulonephritis reduced glomerular and secondary tubulointerstitial damage. METHODS: Using this model, we now assessed the effects of CR002 (n=15) vs irrelevant control IgG (n=17) administered on days 17, 28 and 35 after disease induction, i.e. after acute glomerular damage had subsided. RESULTS: In vitro, CR002 inhibited the PDGF-D- but not the PDGF-B-induced proliferation of rat renal fibroblasts. Following the first CR002 injection on day 17, exposure to therapeutic levels was maintained until day 49. Proteinuria in the CR002-treated group was transiently reduced between days 49 and 77 (-19 to -23% in comparison with the controls; P<0.05). On day 100, CR002 treatment reduced the number of rats that had doubled their serum creatinine (CR002: 40 vs controls: 71%; P<0.05). Compared with controls, the CR002 animals, on day 100, significantly lowered glomerular expression of vimentin and collagens as well as tubulointerstitial damage scores, interstitial fibrosis, vimentin and cortical PDGF-D mRNA levels. CONCLUSIONS: PDGF-D antagonism, even after the phase of acute glomerular damage, exerts beneficial effects on the course of tubulointerstitial damage, i.e. the final common pathway of most renal diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Glomerulonephritis/drug therapy , Kidney Tubules/pathology , Lymphokines/antagonists & inhibitors , Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Disease Progression , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/pathology , Glomerulonephritis/complications , Glomerulonephritis/pathology , Humans , Immunoglobulin G/administration & dosage , Kidney Tubules/drug effects , Lymphokines/immunology , Lymphokines/pharmacology , Male , Platelet-Derived Growth Factor/immunology , Platelet-Derived Growth Factor/pharmacology , Proteinuria/physiopathology , Proteinuria/prevention & control , Rats , Rats, Wistar , Renal Insufficiency/physiopathology , Renal Insufficiency/prevention & control , Vimentin/metabolism
9.
Mol Cancer Ther ; 5(8): 2086-95, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928830

ABSTRACT

Histone deacetylase inhibitors represent a promising new class of anticancer agents. In the current investigation, we examined the activity of PXD101, a potent histone deacetylase inhibitor, used alone or in combination with clinically relevant chemotherapeutics (docetaxel, paclitaxel, and carboplatin), in preclinical in vitro and in vivo models of ovarian cancer. In vitro activity was examined in ovarian cancer and multidrug-resistant cell lines grown in monolayer culture, and in primary clinical ovarian cancer specimens grown in three-dimensional organoid culture. PXD101 was found to inhibit in vitro cancer cell growth at sub- to low micromolar IC(50) potency, exhibited synergistic activity when used in combination with relevant chemotherapeutics, and effectively inhibited the growth of multidrug-resistant cells. In vivo, PXD101 displayed single-agent antitumor activity on human A2780 ovarian cancer s.c. xenografts which was enhanced via combination therapy with carboplatin. In support of these findings, PXD101 was shown to increase the acetylation of alpha-tubulin induced by docetaxel and the phosphorylation of H2AX induced by carboplatin. Taken together, these results support the clinical evaluation of PXD101 used alone or in combination therapy for the treatment of ovarian cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Ovarian Neoplasms/drug therapy , Acetylation , Animals , Antineoplastic Combined Chemotherapy Protocols , Carboplatin/administration & dosage , Cell Line, Tumor , Docetaxel , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Histones/drug effects , Histones/metabolism , Humans , Hydroxamic Acids/administration & dosage , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Phosphorylation , Sulfonamides , Taxoids/administration & dosage , Taxoids/pharmacology , Tubulin/drug effects , Tubulin/metabolism
10.
Clin Cancer Res ; 12(4): 1373-82, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489096

ABSTRACT

PURPOSE: Advanced melanoma is a highly drug-refractory neoplasm representing a significant unmet medical need. We sought to identify melanoma-associated cell surface molecules and to develop as well as preclinically test immunotherapeutic reagents designed to exploit such targets. EXPERIMENTAL DESIGN AND RESULTS: By transcript profiling, we identified glycoprotein NMB (GPNMB) as a gene that is expressed by most metastatic melanoma samples examined. GPNMB is predicted to be a transmembrane protein, thus making it a potential immunotherapeutic target in the treatment of this disease. A fully human monoclonal antibody, designated CR011, was generated to the extracellular domain of GPNMB and characterized for growth-inhibitory activity against melanoma. The CR011 monoclonal antibody showed surface staining of most melanoma cell lines by flow cytometry and reacted with a majority of metastatic melanoma specimens by immunohistochemistry. CR011 alone did not inhibit the growth of melanoma cells. However, when linked to the cytotoxic agent monomethylauristatin E (MMAE) to generate the CR011-vcMMAE antibody-drug conjugate, this reagent now potently and specifically inhibited the growth of GPNMB-positive melanoma cells in vitro. Ectopic overexpression and small interfering RNA transfection studies showed that GPNMB expression is both necessary and sufficient for sensitivity to low concentrations of CR011-vcMMAE. In a melanoma xenograft model, CR011-vcMMAE induced significant dose-proportional antitumor effects, including complete regressions, at doses as low as 1.25 mg/kg. CONCLUSION: These preclinical results support the continued evaluation of CR011-vcMMAE for the treatment of melanoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Melanoma, Experimental/drug therapy , Membrane Glycoproteins/immunology , Oligopeptides/therapeutic use , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoconjugates/pharmacology , Immunohistochemistry , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Oligopeptides/chemistry , Oligopeptides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Outcome , Xenograft Model Antitumor Assays/methods
11.
Biochem Biophys Res Commun ; 333(2): 308-15, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15950186

ABSTRACT

The process of angiogenesis plays a pivotal role in embryogenesis, wound healing, and tumorigenesis through the growth of new blood vessels from pre-existing vasculature. Among the angiogenic factors recently identified as specific for vascular endothelium are the angiopoietins. In depth characterization of the angiopoietins has allowed investigators to better understand the molecular basis of blood vessel formation and vascular endothelial cell function. In this review, we describe angiopoietins and related family members, with particular emphasis on a recently identified protein known as angioarrestin. Our investigations clearly demonstrate that angioarrestin is an anti-angiogenic molecule. The effects of angioarrestin on tumor cell progression and specific aspects of the angiogenic cascade in in vitro models are further discussed.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Angiopoietin-Like Protein 1 , Angiopoietin-like Proteins , Angiopoietins , Animals , Humans
12.
Cancer Biol Ther ; 4(6): 659-68, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15917651

ABSTRACT

The Semaphorins are a large family of transmembrane, GPI-anchored and secreted proteins that play an important role in neuronal and endothelial cell guidance. A human gene related to the class 6 Semaphorin family, Semaphorin 6A-1 (Sema 6A-1) was identified by homology-based genomic mining. Recent implication of Sema 3 family members in tumor angiogenesis and our expression analysis of Sema 6A-1 suggested that class 6 Semaphorin might effect tumor neovascularization. The mRNA expression of Sema 6A-1 was elevated in several renal tumor tissue samples relative to adjacent nontumor tissue samples from the same patient. Sema 6A-1 transcript was also expressed in the majority of renal clear cell carcinoma (RCC) cell lines and to a lesser extent in endothelial cells. To test the role of Sema 6A-1 in tumor angiogenesis, we engineered, expressed and purified the Sema 6A-1 soluble extracellular domain (Sema-ECD). The purified Sema-ECD was screened in a variety of endothelial cell-based assays both in vitro and in vivo. In vitro, Sema-ECD blocked VEGF-mediated endothelial cell migration. These effects were explained in part by our observation in endothelial cells that Sema-ECD inhibited VEGF-mediated Src, FAK and ERK phosphorylation. In vivo, mouse Matrigel assays demonstrated that the intraperitoneal administration of recombinant Sema-ECD inhibited both bFGF/VEGF and tumor cell line-induced neovascularization. These findings reveal a novel therapeutic utility for Sema 6A-1 (Sema-ECD) as an inhibitor of growth factor as well as tumor-induced angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/blood supply , Fibroblast Growth Factor 2/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , Semaphorins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adenocarcinoma, Clear Cell/blood supply , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/therapy , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , Cell Movement/drug effects , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblast Growth Factor 2/pharmacology , Focal Adhesion Kinase 1/metabolism , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/therapy , Laminin/metabolism , Mice , Mice, Nude , Phosphorylation , Protein Structure, Tertiary , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Semaphorins/genetics , Vascular Endothelial Growth Factor A/pharmacology , src-Family Kinases/metabolism
13.
Curr Med Chem Anticancer Agents ; 5(2): 115-30, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777219

ABSTRACT

The inhibition of angiogenesis is an emerging therapeutic strategy for cancer treatment. In contrast to conventional therapies, anti-angiogenic therapies primarily target tumor-associated endothelial cells which serve as a lifeline for tumor growth, progression and metastasis. By blocking the supply of essential nutrients and the removal of metabolites, anti-angiogenic therapies aim to delay both primary and metastastic tumor growth while overcoming the inherent cytotoxicities of classical chemotherapies. Indeed, tumor-related angiogenesis is a multi-step process initiated by a cascade of proangiogenic factors secreted from both the tumor and host tissues. These intricate processes involve a close interaction of tumor and associated endothelial cells as well as an intimate communication between proliferating endothelial cells, stromal cells and extracellular matrix components. Inhibition of these proangiogenic mechanisms has become a major challenge for the development of anti-cancer treatment modalities. In this regard, anti-angiogenic therapies embody a potentially powerful adjunct to traditional cancer therapies. In this review, we provide an overview of traditional anti-cancer drugs and discuss the fundamentals of anti-angiogenic therapies. While presenting the salient features of the anti-angiogenic agents targeting the individual phases of angiogenesis, we highlight the potential for specific agent development as novel anti-angiogenic therapeutics. Finally, we present and summarize emerging angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Angiogenesis Inhibitors/metabolism , Animals , Drug Design , Humans , Models, Biological , Molecular Conformation , Neoplasms/metabolism , Structure-Activity Relationship
14.
Clin Cancer Res ; 9(9): 3454-61, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12960137

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the activity of CG53135 (FGF-20), a protein with in vitro mitogenic activity on epithelial and mesenchymal cells, in two in vivo models of oral mucositis (OM). EXPERIMENTAL DESIGN: Radiation or concomitant chemotherapy/radiation-induced OM was elicited in hamsters. Activity of CG53135 was assessed at different doses and regimens in the models. Bromodeoxyuridine (BrdUrd) incorporation and pharmacokinetic studies were also performed to correlate in vivo activity of CG53135 with exposure. RESULTS: In the hamster radiation model, administration of CG53135 (600 or 1200 micro g/day, i.p.) on days 3-15 resulted in a statistically significant (P < 0.001) reduction in days spent with severe mucositis. CG53135 administered at 12 mg/kg, i.p. (days 1-2 or 1-8) in the concomitant chemotherapy/radiation model resulted in a statistically significant (P < 0.001) reduction in severe mucositis. Maximal BrdUrd incorporation was observed in cheek pouch and jejunal tissues at 8 h, and peak plasma levels of CG53135 were reached 1 h after administration. CONCLUSIONS: CG53135 demonstrates potent, regimen-dependent activity in hamster models of OM. The activity was regimen dependent. BrdUrd incorporation studies confirmed that CG53135 had proliferative activity in vivo with a favorable pharmacokinetic profile. Based in part on work described herein, CG53135 has received approval from the United States Food and Drug Administration to be evaluated in a Phase I clinical trial of cancer patients at risk for developing OM.


Subject(s)
Fibroblast Growth Factors/therapeutic use , Mouth Mucosa/pathology , Radiation Injuries/therapy , Animals , Bromodeoxyuridine/pharmacology , Cheek/pathology , Coloring Agents/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Fluorouracil/pharmacology , Humans , Jejunum/metabolism , Male , Mesocricetus , Mucous Membrane/drug effects , Mucous Membrane/radiation effects , Recombinant Proteins/therapeutic use , Stomatitis/chemically induced , Stomatitis/drug therapy , Time Factors
15.
J Am Soc Nephrol ; 14(9): 2237-47, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937299

ABSTRACT

PDGF-B is of central importance in mesangioproliferative diseases. PDGF-D, a new PDGF isoform, like PDGF-B, signals through the PDGF betabeta-receptor. The present study first determined that PDGF-D is mitogenic for rat mesangial cells and is not inhibited by a PDGF-B antagonist. Low levels of PDGF-D mRNA were detected in normal rat glomeruli. After induction of mesangioproliferative nephritis in rats by anti-Thy 1.1 mAb, glomerular PDGF-D mRNA and protein expression increased significantly from days 4 to 9 in comparison with nonnephritic rats. Peak expression of PDGF-D mRNA occurred 2 d later than peak PDGF-B mRNA expression. In addition, PDGF-D serum levels increased significantly in the nephritic animals on day 7. For investigating the functional role of PDGF-D, neutralizing fully human mAb were generated using the XenoMouse technology. Rats with anti-Thy 1.1-induced nephritis were treated on days 3 and 5 with different amounts of a fully human PDGF-DD-specific neutralizing mAb (CR002), equal amounts of irrelevant control mAb, or PBS by intraperitoneal injection. Specific antagonism of PDGF-D led to a dose-dependent (up to 67%) reduction of glomerular cell proliferation. As judged by double immunostaining for 5-bromo-2'-deoxyuridine and alpha-smooth muscle actin, glomerular mesangial cell proliferation was reduced by up to 57%. Reduction of glomerular cell proliferation in the rats that received CR002 was not associated with reduced glomerular expression of PDGF-B mRNA. PDGF-D antagonism also led to reduced glomerular infiltration of monocytes/macrophages (day 5) and reduced accumulation of fibronectin (day 8). In contrast, no effect was noted in normal rats that received an injection of CR002. These data show that PDGF-D is overexpressed in mesangioproliferative states and can act as an auto-, para-, or even endocrine glomerular cell mitogen, indicating that antagonism of PDGF-D may represent a novel therapeutic approach to mesangioproliferative glomerulonephritides.


Subject(s)
Antibodies, Monoclonal/immunology , Glomerular Mesangium/metabolism , Glomerulonephritis, Membranoproliferative/metabolism , Lymphokines , Platelet-Derived Growth Factor/metabolism , Animals , Cell Division/physiology , Cells, Cultured , Down-Regulation , Glomerulonephritis, Membranoproliferative/immunology , Humans , Mice , Platelet-Derived Growth Factor/immunology , Rats , Up-Regulation
16.
Expert Opin Ther Targets ; 6(4): 469-82, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12223061

ABSTRACT

The fibroblast growth factor (FGF) family of signalling molecules and its receptors (FGFRs) contribute to normal developmental and physiological processes. However, the subversion of this powerful growth stimulatory pathway has been implicated in the generation of a variety of pathological conditions. This review focuses on the role of FGF/FGFRs in cancer. The case will be made that this signalling pathway is associated with and functionally important for the growth of some human tumours. As such, FGF/FGFRs can be viewed as rational therapeutic oncology targets and strategies used to inhibit these molecules are discussed. The therapeutic exploitation of tumour-associated FGFR expression to deliver toxins or antiproliferative signals to tumour cells is also reviewed, as is the use of FGFs as protein therapeutics to alleviate the side effects of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Fibroblast Growth Factors/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Fibroblast Growth Factor/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Cell Division/drug effects , Drug Carriers , Drug Delivery Systems , Drug Screening Assays, Antitumor , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/physiology , Fibroblast Growth Factors/therapeutic use , Humans , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Neoplasms/physiopathology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction/drug effects
17.
Cancer Res ; 62(13): 3834-41, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12097297

ABSTRACT

The angiopoietins comprise a family of proteins that have pro or antiangiogenic activities. Through a proprietary technology designed to identify transcripts of all expressed genes, we isolated a cDNA encoding an angiopoietin-related protein that we designate angioarrestin. The mRNA expression profile of angioarrestin was striking in that it was down-regulated in many tumor tissues when compared with adjacent nontumor tissue, suggesting a role for this protein in tumor inhibition. To test this hypothesis, we ectopically expressed angioarrestin in HT1080 tumor cells and measured pulmonary tumor nodule formation in nude mice. HT1080 cells expressing angioarrestin showed a marked reduction in the number and size of tumor nodules. In vitro, the recombinant protein was systematically tested in a number of endothelial cell assays and found to block critical processes involved in the angiogenic cascade, such as vascular endothelial growth factor/basic fibroblast growth factor-mediated endothelial cell proliferation, migration, tubular network formation, and adhesion to extracellular matrix proteins. These findings reveal a novel function for angioarrestin as an angiogenesis inhibitor and indicate that the molecule may be a potential cancer therapeutic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Proteins/pharmacology , 3T3 Cells , Amino Acid Sequence , Angiopoietin-Like Protein 1 , Angiopoietin-like Proteins , Angiopoietins , Animals , Base Sequence , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix Proteins/metabolism , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
18.
Cancer Res ; 62(9): 2468-73, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980634

ABSTRACT

Platelet-derived growth factor (PDGF) has been directly implicated in developmental and physiological processes, as well as in human cancer and other proliferative disorders. We have recently isolated and characterized a novel protease-activated member of the PDGF family, PDGF D. PDGF D has been shown to be proliferative for cells of mesenchymal origin, signaling through PDGF receptors. Comprehensive and systematic PDGF D transcript analysis revealed expression in many cell lines derived from ovarian, renal, and lung cancers, as well as from astrocytomas and medulloblastomas. beta PDGF receptor profiling further suggested autocrine signaling in several brain tumor cell lines. PDGF D transforming ability and tumor formation in SCID mice was further demonstrated. Exploiting a sensitive PDGF D sandwich ELISA using fully human monoclonal antibodies, PDGF D was detected at elevated levels in the sera of ovarian, renal, lung, and brain cancer patients. Immunohistochemical analysis confirmed PDGF D localization to ovarian and lung tumor tissues. Together, these data demonstrate that PDGF D plays a role in certain human cancers.


Subject(s)
Neoplasms/metabolism , Platelet-Derived Growth Factor/physiology , 3T3 Cells , Animals , Cell Transformation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, SCID , Neoplasms/blood , Neoplasms/pathology , Phosphorylation , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/physiology , Signal Transduction/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...