Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 13(1): 2304963, 2024.
Article in English | MEDLINE | ID: mdl-38235317

ABSTRACT

Approximately two-thirds of hepatocellular carcinoma (HCC) is considered a "cold tumor" characterized by few tumor-infiltrating T cells and an abundance of immunosuppressive cells. Cilengitide, an integrin αvß3 inhibitor, has failed in clinical trials as a potential anticancer drug. This failure implies that integrin αvß3 may play an important role in immune cells. However, the expression and potential role of integrin αvß3 in T cells of HCC patients remain unknown. Here, we established two HCC models and found that cilengitide had a dual effect on the HCC microenvironment by exerting both antitumor effect and immunosuppressive effect on T cells. This may partly explain the failure of cilengitide in clinical trials. In clinical specimens, HCC-infiltrating T cells exhibited deficient expression and activation of integrin ß3, which was associated with poor T-cell infiltration into tumors. Additionally, integrin ß3 functioned as a positive immunomodulatory molecule to facilitate T-cell infiltration and T helper 1-type immune response in vitro. Furthermore, T cells and platelet-derived microparticles (PMPs) co-culture assay revealed that PMPs adoptively transferred integrin ß3 to T cells and positively regulated T cell immune response. This process was mediated by clathrin-dependent endocytosis and macropinocytosis. Our data demonstrate that integrin ß3 deficiency on HCC-infiltrating T cells may be involved in shaping the immunosuppressive tumor microenvironment. PMPs transfer integrin ß3 to T cells and positively regulate T cell immune response, which may provide a new insight into immune therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell-Derived Microparticles , Liver Neoplasms , Humans , Integrin beta3/metabolism , Integrin beta3/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Integrin alphaVbeta3/therapeutic use , Liver Neoplasms/drug therapy , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , T-Lymphocytes , Tumor Microenvironment
2.
Cornea ; 43(3): 378-386, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38015979

ABSTRACT

PURPOSE: The purpose of this study was to extensively evaluate the efficacy of integrin αvß3 antagonists for the treatment of experimental dry eye (EDE). METHODS: Vitronectin, an αvß3 ligand, was used to induce tumor necrosis factor-α gene expression in human THP-1 macrophages. To induce EDE, C57BL/6 mice were housed in a low-humidity controlled environment chamber and injected subcutaneously with scopolamine for 7 days. Subsequently, αvß3 antagonists, including RGDfD, c(RGDfD), c(RGDiD), c(RGDfK), ATN-161, SB273005, and cilengitide, were administered topically to EDE animals under controlled environment chamber conditions. Corneal epithelial damage in EDE was assessed by fluorescein staining. The density of conjunctival goblet cells and secretion of tears was measured by period acid-Schiff staining and phenol red-impregnated cotton threads, respectively. Inflammation markers, including tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, IL-17A, and metalloproteinase (MMP)-9, in the pooled cornea and conjunctiva tissues were examined by real-time polymerase chain reaction. RESULTS: The inhibitory effects of αvß3 antagonists on the vitronectin-induced tumor necrosis factor-α gene expression and integrin-mediated inflammatory signaling were validated in THP-1 macrophages. αvß3 antagonists ameliorated the impairment of the corneal epithelial barrier with varying therapeutic efficacies, compared with vehicle-treated mice. c(RGDfD) and c(RGDiD) significantly protected against goblet cell loss, tear reduction, and proinflammatory gene expression in EDE. CONCLUSIONS: Topical applications of αvß3 antagonists yield therapeutic benefits in EDE by promoting corneal epithelial defect healing and reducing inflammation. Antagonistic targeting αvß3 may be a novel promising strategy to treat patients with dry eye disease.


Subject(s)
Dry Eye Syndromes , Integrin alphaVbeta3 , Humans , Animals , Mice , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/therapeutic use , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vitronectin/metabolism , Vitronectin/pharmacology , Vitronectin/therapeutic use , Mice, Inbred C57BL , Dry Eye Syndromes/metabolism , Tears/metabolism , Conjunctiva/pathology , Cornea/pathology , Inflammation/metabolism , Disease Models, Animal
3.
Acta Pharmacol Sin ; 44(4): 897-912, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36280689

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) drugs have revolutionized the treatment of neovascular eye diseases, but responses are incomplete in some patients. Recent evidence shows that integrins are involved in the pathogenesis of neovascular age-related macular degeneration and diabetic retinopathy. JP1, derived from an optimized seven-amino-acid fragment of JWA protein, is a polypeptide specifically targeting integrin αVß3. In this study we evaluated the efficacy of JP1 on laser-induced choroidal neovascularization (CNV) and retinal vascular leakage. CNV mice received a single intravitreal (IVT) injection of JP1 (10, 20, 40 µg) or ranibizumab (RBZ, 10 µg). We showed that JP1 injection dose-dependently inhibited laser-induced CNV; the effect of RBZ was comparable to that of 20 µg JP1; a combined IVT injection of JP1 (20 µg) and RBZ (5 µg) exerted a synergistic effect on CNV. In the 3rd month after streptozotocin injection, diabetic mice receiving IVT injection of JP1 (40 µg) or RBZ (10 µg) once a week for 4 weeks showed significantly suppressed retinal vascular leakage. In both in vivo and in vitro experiments, JP1 counteracted oxidative stress and inflammation via inhibiting ROS/NF-κB signaling in microglial cells, and angiogenesis via modulating MEK1/2-SP1-integrin αVß3 and TRIM25-SP1-MMP2 axes in vascular endothelial cells. In addition, intraperitoneal injection of JP1 (1, 5 or 10 mg) once every other day for 3 times also dose-dependently inhibited CNV. After intraperitoneal injection of FITC-labeled JP1 (FITC-JP1) or FITC in laser-induced CNV mice, the fluorescence intensity in the CNV lesion was markedly increased in FITC-JP1 group, compared with that in FITC group, confirming that JP1 could penetrate the blood-retinal barrier to target CNV lesion. We conclude that JP1 can be used to design novel CNV-targeting therapeutic agents that may replace current invasive intraocular injections.


Subject(s)
Choroidal Neovascularization , Diabetes Mellitus, Experimental , Diabetic Retinopathy , Animals , Mice , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Disease Models, Animal , Endothelial Cells/metabolism , Fluorescein-5-isothiocyanate/therapeutic use , Integrin alphaVbeta3/therapeutic use , Peptides/therapeutic use
4.
Acta Neuropathol Commun ; 10(1): 151, 2022 10 23.
Article in English | MEDLINE | ID: mdl-36274147

ABSTRACT

RATIONALE: Glioblastoma multiforme (GBM) is a primary brain tumor with poor prognosis. The U.S. food and drug administration approved the use of the anti-VEGF antibody bevacizumab in recurrent GBM. However, resistance to this treatment is frequent and fails to enhance the overall survival of patients. In this study, we aimed to identify novel mechanism(s) responsible for bevacizumab-resistance in CD146-positive glioblastoma. METHODS: The study was performed using sera from GBM patients and human GBM cell lines in culture or xenografted in nude mice. RESULTS: We found that an increase in sCD146 concentration in sera of GBM patients after the first cycle of bevacizumab treatment was significantly associated with poor progression free survival and shorter overall survival. Accordingly, in vitro treatment of CD146-positive glioblastoma cells with bevacizumab led to a high sCD146 secretion, inducing cell invasion. These effects were mediated through integrin αvß3 and were blocked by mucizumab, a novel humanized anti-sCD146 antibody. In vivo, the combination of bevacizumab with mucizumab impeded CD146 + glioblastoma growth and reduced tumor cell dissemination to an extent significantly higher than that observed with bevacizumab alone. CONCLUSION: We propose sCD146 to be 1/ an early biomarker to predict and 2/ a potential target to prevent bevacizumab resistance in patients with glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Mice , Animals , Humans , Glioblastoma/pathology , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , CD146 Antigen/metabolism , Mice, Nude , Integrin alphaVbeta3/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Biomarkers , Brain Neoplasms/pathology
5.
Cancer Res Treat ; 54(3): 767-781, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34607394

ABSTRACT

PURPOSE: Heat shock protein-90 (HSP90) remains an important cancer target because of its involvement in multiple oncogenic protein pathways and biologic processes. Although many HSP90 inhibitors have been tested in the treatment of KRAS-mutant non-small cell lung cancer (NSCLC), most, including AUY922, have failed due to toxic effects and resistance generation, even though a modest efficacy has been observed for these drugs in clinical trials. In our present study, we investigated the novel mechanism of resistance to AUY922 to explore possible avenues of overcoming and want to provide some insights that may assist with the future development of successful next-generation HSP90 inhibitors. MATERIALS AND METHODS: We established two AUY922-resistant KRAS-mutated NSCLC cells and conducted RNA sequencing to identify novel resistance biomarker. RESULTS: We identified novel two resistance biomarkers. We observed that both integrin Av (ITGAv) and ß3 (ITGB3) induce AUY922-resistance via focal adhesion kinase (FAK) activation, as well as an epithelial-mesenchymal transition, in both in vitro and in vivo xenograft model. mRNAs of both ITGAv and ITGB3 were also found to be elevated in a patient who had shown acquired resistance in a clinical trial of AUY922. ITGAv was induced by miR-142 downregulation, and ITGB3 was increased by miR-150 downregulation during the development of AUY922-resistance. Therefore, miR-150 and miR-142 overexpression effectively inhibited ITGAvB3-dependent FAK activation, restoring sensitivity to AUY922. CONCLUSION: The synergistic co-targeting of FAK and HSP90 attenuated the growth of ITGAvB3-induced AUY922-resistant KRAS-mutated NSCLC cells in vitro and in vivo, suggesting that this combination may overcome acquired AUY922-resistance in KRAS-mutant NSCLC.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
6.
J Immunol Res ; 2021: 9958239, 2021.
Article in English | MEDLINE | ID: mdl-33880384

ABSTRACT

METHODS: Superparamagnetic iron oxide nanoclusters (SPIOCs) were located within the core, which resulted in high photothermal conversion and outstanding generation of reactive oxygen species (ROS). The shell consisted of a human serum albumin- (HSA-) paclitaxel (PTX) layer, which extended the blood circulation time and ensured the effectiveness of the chemotherapy. Arg-Gly-Asp peptides (RGD) were linked to the naked cysteine moieties in HSA to promote the specific targeting of human glioma U87 cells by α v ß 3 integrins. Continuous near-infrared light irradiation triggered and promoted the synergistic chemo/CDT therapy through the photothermal effect. RESULTS: Our SPIOCs@HSA-RGD nanoplatform showed well biocompatibility and could target glioma specifically. Photothermal conversion and ROS burst were detected after continuous 808 nm light irradiation, and a significant antitumor effect was achieved. CONCLUSION: Experimental in vitro and in vivo evaluations showed that our photothermal-mediated chemo/CDT therapy could efficiently inhibit tumor growth and is therefore promising for cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/methods , Glioma/therapy , Integrin alphaVbeta3/therapeutic use , Oligopeptides/therapeutic use , Paclitaxel/therapeutic use , Theranostic Nanomedicine/methods , Animals , Cell Growth Processes , Cell Line, Tumor , Drug Synergism , Humans , Infrared Rays , Integrin alphaVbeta3/metabolism , Magnetic Iron Oxide Nanoparticles/administration & dosage , Magnetic Iron Oxide Nanoparticles/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/chemistry , Paclitaxel/chemistry , Rats , Reactive Oxygen Species/metabolism , Serum Albumin, Human/chemistry , Xenograft Model Antitumor Assays
7.
J Biomed Sci ; 28(1): 24, 2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33827580

ABSTRACT

Thyroid hormone analogues-particularly, L-thyroxine (T4) has been shown to be relevant to the functions of a variety of cancers. Integrin αvß3 is a plasma membrane structural protein linked to signal transduction pathways that are critical to cancer cell proliferation and metastasis. Thyroid hormones, T4 and to a less extend T3 bind cell surface integrin αvß3, to stimulate the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway to stimulate cancer cell growth. Thyroid hormone analogues also engage in crosstalk with the epidermal growth factor receptor (EGFR)-Ras pathway. EGFR signal generation and, downstream, transduction of Ras/Raf pathway signals contribute importantly to tumor cell progression. Mutated Ras oncogenes contribute to chemoresistance in colorectal carcinoma (CRC); chemoresistance may depend in part on the activity of ERK1/2 pathway. In this review, we evaluate the contribution of thyroxine interacting with integrin αvß3 and crosstalking with EGFR/Ras signaling pathway non-genomically in CRC proliferation. Tetraiodothyroacetic acid (tetrac), the deaminated analogue of T4, and its nano-derivative, NDAT, have anticancer functions, with effectiveness against CRC and other tumors. In Ras-mutant CRC cells, tetrac derivatives may overcome chemoresistance to other drugs via actions initiated at integrin αvß3 and involving, downstream, the EGFR-Ras signaling pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , Integrin alphaVbeta3/therapeutic use , Signal Transduction , Thyroxine/therapeutic use , Genes, erbB-1 , Genes, ras , Humans
8.
Oncogene ; 38(25): 5050-5061, 2019 06.
Article in English | MEDLINE | ID: mdl-30872792

ABSTRACT

Cancer cells frequently exhibit higher levels of reactive oxygen species (ROS) than normal cells and when ROS levels increase beyond a cellular tolerability threshold, cancer cell death is enhanced. The mitochondrial dihydrolipoamide dehydrogenase (DLDH) is an enzyme which produces ROS in association with its oxidoreductive activity and may be thus utilized as an exogenous anticancer agent. As cancer cells often overexpress integrins that recognize RGD-containing proteins, we have bioengineered the human DLDH with RGD motifs (DLDHRGD) for integrin-mediated drug delivery. The modified protein fully retained its enzyme activity and ROS-production capability. DLDHRGD uptake by cells was shown to depend on the presence of cell-associated integrin αvß3, as comparatively demonstrated with normal kidney cells (HEK293) transfected with either ß1 (αvß1 positive) or ß3 integrins (αvß3 positive). The interaction with ß3 integrins was shown to be competitively inhibited by an RGD peptide. In mice melanoma cells (B16F10), which highly express an endogenous αvß3 integrin, fast cellular uptake of DLDHRGD which resulted in cell number reduction, apoptosis induction, and a parallel intracellular ROS production was shown. Similar results were obtained with additional human melanoma cell models (A375, WM3314, and WM3682). In contrast, HEK293ß3 cells remained intact following DLDHRGD uptake. The high pharmacological safety profile of DLDHRGD has been observed by several modes of administrations in BALB/C or C57Bl/6 mouse strains. Treatments with DLDHRGD in a subcutaneous melanoma mice model resulted in significant tumor inhibition. Our study demonstrated, in vitro and in vivo, the development of a unique platform, which targets cancer cells via integrin-mediated drug delivery of an exogenous ROS-generating drug.


Subject(s)
Dihydrolipoamide Dehydrogenase/administration & dosage , Drug Delivery Systems , Integrin alphaVbeta3/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Dihydrolipoamide Dehydrogenase/chemistry , Dihydrolipoamide Dehydrogenase/metabolism , Female , HEK293 Cells , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/physiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/metabolism , Oligopeptides/chemistry , Oxidation-Reduction , Xenograft Model Antitumor Assays
9.
J Control Release ; 224: 217-228, 2016 Feb 28.
Article in English | MEDLINE | ID: mdl-26739551

ABSTRACT

Multifunctional near-infrared (NIR) nanoparticles demonstrate great potential in tumor theranostic applications. To achieve the sensitive detection and effective phototherapy in the early stage of tumor genesis, it is highly desirable to improve the targeting of NIR theranostic agents to biomarkers and to enhance their accumulation in tumor. Here we report a novel targeted multifunctional theranostic nanoparticle, internalized RGD (iRGD)-modified indocyanine green (ICG) liposomes (iRGD-ICG-LPs), for molecular imaging-guided photothermal therapy (PTT) and photodynamic therapy (PDT) therapy against breast tumor. The iRGD peptides with high affinity to αvß3 integrin and effective tumor-internalized property were firstly used to synthesize iRGD-PEG2000-DSPE lipopeptides, which were further utilized to fabricate the targeted ICG liposomes. The results indicated that iRGD-ICG-LPs exhibited excellent stability and could provide an accurate and sensitive detection of breast tumor through NIR fluorescence molecular imaging. We further employed this nanoparticle for tumor theranostic application, demonstrating significantly higher tumor accumulation and tumor inhibition efficacy through PTT/PDT effects. Histological analysis further revealed much more apoptotic cells, confirming the advantageous anti-tumor effect of iRGD-ICG-LPs over non-targeted ICG-LPs. Notably, the targeting therapy mediated by iRGD provides almost equivalent anti-tumor efficacy at a 12.5-fold lower drug dose than that by monoclonal antibody, and no tumor recurrence and obvious treatment-induced toxicity were observed in our study. Our study provides a promising strategy to realize the sensitive detection and effective treatment of tumors by integrating molecular imaging into PTT/PDT therapy.


Subject(s)
Coloring Agents/therapeutic use , Indocyanine Green/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Molecular Imaging/methods , Oligopeptides/therapeutic use , Photochemotherapy/methods , Animals , Apoptosis/drug effects , Cell Line , Cell Survival , Coloring Agents/pharmacokinetics , Drug Delivery Systems/methods , Female , Indocyanine Green/pharmacokinetics , Integrin alphaVbeta3/administration & dosage , Integrin alphaVbeta3/therapeutic use , Liposomes , Mice , Mice, Inbred BALB C , Nanoparticles , Polyethylene Glycols/chemistry , Spectroscopy, Near-Infrared , Theranostic Nanomedicine , Tissue Distribution
10.
J. physiol. biochem ; 65(2): 147-156, abr.-jun. 2009. graf
Article in English | IBECS | ID: ibc-75575

ABSTRACT

Thrombotic diseases or fatalities have been reported to occasionally occur underconditions of hypergravity although the mechanism is still unclear. To investigate theeffect of hypergravity on platelets that are the primary players in thrombus formation,platelet rich plasma (PRP) or washed platelets were exposed to hypergravity at8 G for 15 minutes. No platelet aggregation was induced by 8 G alone, whereas ristocetinor collagen-induced platelet aggregation was significantly increased. Thenumber of platelets adherent to immobilized fibrinogen and the area of plateletsspreading on von Willbrand factor (VWF) matrix were increased simultaneously.Flow cytometry assay indicated that integrin áIIbâ3 was partially activated in 8 Gexposedplatelets, but there was no significant difference in P-selectin surface expressionbetween platelets treated with 8 G and 1 G control. The results indicate thathypergravity leads to human platelet hyperactivity, but fails to incur essential plateletactivation events, suggesting a novel mechanism for thrombotic diseases occurringunder hypergravitional conditions(AU)


Subject(s)
Animals , Rats , Hypergravity , Hyperkinesis , Blood Platelets , Thrombosis , Thrombosis/epidemiology , Thrombosis/etiology , Thrombosis/therapy , Integrin alphaVbeta3 , Integrin alphaVbeta3/therapeutic use , Ristocetin
11.
Nat Med ; 15(4): 392-400, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19305413

ABSTRACT

Inhibitors of alpha(v)beta(3) and alpha(v)beta(5) integrin have entered clinical trials as antiangiogenic agents for cancer treatment but generally have been unsuccessful. Here we present in vivo evidence that low (nanomolar) concentrations of RGD-mimetic alpha(v)beta(3) and alpha(v)beta(5) inhibitors can paradoxically stimulate tumor growth and tumor angiogenesis. We show that low concentrations of these inhibitors promote VEGF-mediated angiogenesis by altering alpha(v)beta(3) integrin and vascular endothelial growth factor receptor-2 trafficking, thereby promoting endothelial cell migration to VEGF. The proangiogenic effects of low concentrations of RGD-mimetic integrin inhibitors could compromise their efficacy as anticancer agents and have major implications for the use of RGD-mimetic compounds in humans.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrin alphaVbeta3/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Receptors, Vitronectin/therapeutic use , Animals , Disease Models, Animal , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Vascular Endothelial Growth Factor A/pharmacology
12.
Ann Oncol ; 18(8): 1400-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17693653

ABSTRACT

BACKGROUND: Cilengitide, an antiangiogenic agent that inhibits the binding of integrins alpha(nu)beta(3) and alpha(nu)beta(5) to the extracellular matrix, was studied at two dose levels in cancer patients to determine the optimal biological dose. PATIENTS AND METHODS: The doses of cilengitide were 600 or 1200 mg/m(2) as a 1-h infusion twice weekly every 28 days. A novel dose escalation scheme was utilized that relied upon the biological activity rate. RESULTS: Twenty patients received 50 courses of cilengitide with no dose-limiting toxic effects. The pharmacokinetic (PK) profile revealed a short elimination half-life of 4 h, supporting twice weekly dosing. Of the six soluble angiogenic molecules assessed, only E-selectin increased significantly from baseline. Analysis of tumor microvessel density and gene expression was not informative due to intrapatient tumor heterogeneity. Although several patients with evaluable tumor biopsy pairs did reveal posttreatment increases in tumor and endothelial cell apoptosis, these results did not reach statistical significance due to the aforementioned heterogeneity. CONCLUSIONS: Cilengitide is a well-tolerated antiangiogenic agent. The biomarkers chosen in this study underscore the difficulty in assessing the biological activity of antiangiogenic agents in the absence of validated biological assays.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrin alphaVbeta3/therapeutic use , Integrins/therapeutic use , Neoplasms/drug therapy , Receptors, Vitronectin/therapeutic use , Snake Venoms/therapeutic use , Angiogenesis Inhibitors/pharmacokinetics , Apoptosis/drug effects , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Gene Expression/drug effects , Humans , In Situ Nick-End Labeling , Neoplasms/blood , Snake Venoms/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...