Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
2.
J Pharm Biomed Anal ; 126: 117-23, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27187764

ABSTRACT

A liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method for the determination of taselisib (GDC-0032, RO5537381) concentrations in human plasma has been developed and validated to support bioanalysis of clinical samples. Solid phase extraction (SPE) was used to extract plasma samples (50µL) and the resulting samples were analyzed using reversed phase chromatography and mass spectrometry coupled with an atmospheric pressure chemical ionization interface. The mass analysis of taselisib was performed using multiple reaction monitoring transitions in positive ionization mode. The method was validated over the calibration curve range 0.400-400ng/mL using linear regression and 1/x(2) weighting. The within-run relative standard deviation (%RSD) ranged from 1.3 to 5.6%, while the between-run %RSD varied from 2.0 to 4.5% for LLOQ, low, medium, medium high and high QCs. The accuracy ranged from 94.7 to 100.3% of nominal for within-run and 96.0-99.0% of nominal for between-run for the same QCs. Extraction recovery of taselisib was between 83.8% and 92.9%. Stability of taselisib was established in human plasma for 977days at -20°C and -70°C and established in sample extracts for 96h when stored at 2 - 8°C. Stable-labeled internal standard was used to minimize matrix effects. Mean single dose pharmacokinetic parameters determined using this method for a phase I/II clinical trial were: Cmax=35.2ng/mL, AUC0-inf=1570ngh/mL, and T1/2=39.3h.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Imidazoles/blood , Oxazepines/blood , Tandem Mass Spectrometry/methods , Antineoplastic Agents/analysis , Area Under Curve , Calibration , Chromatography, Reverse-Phase/methods , Drug Stability , Drug Storage , Half-Life , Humans , Imidazoles/analysis , Linear Models , Oxazepines/analysis , Phosphoinositide-3 Kinase Inhibitors , Reproducibility of Results , Solid Phase Extraction/methods , Temperature , Time Factors
3.
J Pharm Biomed Anal ; 100: 150-156, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165011

ABSTRACT

A liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method for the determination of GDC-0980 (Apitolisib) concentrations in human plasma has been developed and validated to support clinical development. Supported liquid extraction (SLE) was used to extract plasma samples (80µL) and the resulting samples were analyzed using reverse-phase chromatography and mass spectrometry coupled with a turbo-ionspray interface. The mass analysis of GDC-0980 was performed using multiple reaction monitoring (MRM) transitions in positive ionization mode. The method was validated over the calibration curve range 0.0500-25.0ng/mL using linear regression and 1/x(2) weighting. Within-run relative standard deviation (%RSD) ranged from 0.4 to 3.9%, while the between-run %RSD varied from 1.1 to 1.5% for QCs. The accuracy ranged from 96.1% to 106.7% of nominal for within-run and 96.7-106.7% of nominal for between-run at all concentrations including the LLOQ quality control at 0.0500ng/mL. Extraction recovery of GDC-0980 was between 72.4% and 75.5%. Stability of GDC-0980 was established in human plasma for 547 days at -20°C and -70°C and established in reconstituted sample extracts for 146h when stored at 2-8°C. Stable-labeled internal standard was used to minimize matrix effects. Mean pharmacokinetic parameters determined using this method for the day 1 control group in a phase I trial were: Cmax=11.1ng/mL, AUC0-inf=108ngh/mL, and T1/2=13.1h.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/blood , Chromatography, Reverse-Phase , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/blood , Pyrimidines/blood , Spectrometry, Mass, Electrospray Ionization , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tandem Mass Spectrometry , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Calibration , Chromatography, Reverse-Phase/standards , Drug Interactions , Female , Half-Life , Humans , Ketoconazole/administration & dosage , Limit of Detection , Linear Models , Middle Aged , Phosphatidylinositol 3-Kinase/administration & dosage , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/standards , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry/standards , Young Adult
5.
Clin Pharmacol Ther ; 92(2): 203-13, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22739140

ABSTRACT

A majority of the novel orally administered, molecularly targeted anticancer therapies are weak bases that exhibit pH-dependent solubility, and suppression of gastric acidity with acid-reducing agents could impair their absorption. In addition, a majority of cancer patients frequently take acid-reducing agents to alleviate symptoms of gastroesophageal reflux disease, thereby raising the potential for a common but underappreciated drug-drug interaction (DDI) that could decrease the exposure of anticancer medication and result in subsequent failure of therapy. This article is a review of the available clinical literature describing the extent of the interaction between 15 orally administered, small-molecule targeted anticancer therapies and acid-reducing agents. The currently available clinical data suggest that the magnitude of this DDI is largest for compounds whose in vitro solubility varies over the pH range 1-4. This range represents the normal physiological gastric acidity (pH ~1) and gastric acidity while on an acid-reducing agent (pH ~4).


Subject(s)
Anticarcinogenic Agents/therapeutic use , Gastroesophageal Reflux/drug therapy , Neoplasms/drug therapy , Proton Pump Inhibitors/therapeutic use , Anticarcinogenic Agents/pharmacokinetics , Drug Interactions , Gastric Acid , Gastric Acidity Determination , Gastroesophageal Reflux/metabolism , Humans , Hydrogen-Ion Concentration , Intestinal Absorption , Neoplasms/metabolism , Proton Pump Inhibitors/pharmacokinetics , Solubility
6.
J Pharm Biomed Anal ; 61: 1-7, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22169467

ABSTRACT

A solid phase extraction (SPE) liquid chromatographic-tandem mass spectrometry (LC-MS/MS) method for the determination of GDC-0941 concentrations in human plasma has been developed and validated to support clinical development. An Oasis MCX 10mg 96-well SPE plate was used to extract plasma samples (50 µL) and the resulting extracts were analyzed using reverse-phase chromatography and mass spectrometer coupled with a turbo-ionspray interface. The method was validated over the calibration curve range 0.500-500 ng/mL with linear regression and 1/x(2) weighting. Within-run relative standard deviation (%RSD) ranged from 1.5 to 11.5%, while the between-run %RSD varied from 0.0 to 4.4%. The accuracy ranged from 96.0% to 110.0% of nominal for within-run and 98.0% to 108.0% of nominal for between-run at all concentrations including the LLOQ quality control at 0.500 ng/mL. Extraction recovery of GDC-0941 was between 79.0% and 86.2%. Stability of GDC-0941 was established in human plasma for 602 days at -70 °C and 598 days at -20°C, respectively, and established in reconstituted sample extracts for 167 h when stored at room temperature. Internal standard normalized matrix factor was 1.1, demonstrating that the use of the stable-labeled internal standard GDC-0941-d(8) effectively compensated observed matrix effect and resulting in no adverse impact on the quality of the data produced. This assay was used for the determination of GDC-0941 human plasma concentrations over a sufficient time period to determine pharmacokinetic parameters at relevant clinical doses.


Subject(s)
Indazoles/blood , Phosphoinositide-3 Kinase Inhibitors , Solid Phase Extraction/methods , Sulfonamides/blood , Tandem Mass Spectrometry/methods , Chromatography, Liquid/methods , Female , Humans , Male , Phosphatidylinositol 3-Kinase/metabolism , Reproducibility of Results
7.
Clin Pharmacol Ther ; 85(2): 173-81, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18987624

ABSTRACT

The clinical pharmacokinetics and in vitro inhibition of digoxin were examined to predict the P-glycoprotein (P-gp) component of drug-drug interactions. Coadministered drugs (co-meds) in clinical trials (N = 123) resulted in a small, 0.1 is predictive of clinical digoxin interactions (AUC and C(max)).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Digoxin/metabolism , Amiodarone/metabolism , Amiodarone/pharmacology , Caco-2 Cells , Digoxin/pharmacology , Drug Interactions , Humans , Pharmaceutical Preparations/metabolism , Retrospective Studies
8.
Am J Physiol Heart Circ Physiol ; 281(1): H359-70, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406504

ABSTRACT

Vascular smooth muscle cell (SMC) migration and proliferation contribute to intimal hyperplasia, and protein kinase C (PKC) may be required for both events. In this report, we investigated the role of PKC in proliferation and migration of SMC derived from the human saphenous vein. Activation of PKC by phorbol-12,13-dibutyrate (PDBu) or (-)-indolactam [(-)-ILV] increases SMC proliferation. Downregulation of PKC activity by prolonged incubation with phorbol ester or inhibition of PKC with chelerythrine in SMC diminished agonist-stimulated proliferation. In contrast, stimulation of PKC with PDBu or (-)-ILV inhibited basal and agonist-induced SMC chemotaxis. Moreover, downregulation of PKC or inhibition with chelerythrine accentuated migration. We postulated that the inhibitory effect of PKC on SMC chemotaxis was mediated through cAMP-dependent protein kinase (protein kinase A, PKA). In support of this hypothesis, we found that activation of PKC in SMC stimulated PKA activity. The cAMP agonist forskolin significantly inhibited SMC chemotaxis. Furthermore, the inhibitory effect of PKC on SMC chemotaxis was completely reversed by cAMP or PKA inhibitors. In search of the PKC isotype(s) underlying these differential effects of PKC in SMC, we identified eight isotypes expressed in human SMC. Only PKC-alpha, -beta I, -delta, and -epsilon were eliminated by downregulation, suggesting that one or more of these four enzymes facilitate the observed phorbol ester-dependent effects of PKC in SMC. In summary, we found that PKC activation enhances proliferation but inhibits migration of human vascular SMC. These differential effect of PKC on vascular cells appears to be mediated through PKC-alpha, -beta I, -delta, and/or -epsilon.


Subject(s)
Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Protein Kinase C/pharmacology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Humans , Isoenzymes/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism
10.
J Pharmacol Exp Ther ; 296(2): 426-33, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160627

ABSTRACT

The mitogen-activated protein kinase signaling cascade is used by many G protein-coupled receptors to initiate functional events. In this study, activation of the Gq/G11-coupled thromboxane A2 (TxA2) receptor (TP) by the TxA2 mimetic IBOP in ECV304 cells was found to induce extracellular regulated kinase (ERK) phosphorylation and tyrosine phosphorylation of the epidermal growth factor receptor (EGFR), which were inhibited by the TP antagonist SQ29548, the EGFR kinase inhibitor AG1478, the Src family kinase inhibitor PP1, the Gi/o protein inhibitor pertussis toxin (PTX), or the protein kinase C (PKC) inhibitor calphostin C. TP activation also increased Src kinase activity, which was blocked by PTX, PP1, and calphostin C, but not by AG1478, indicating that Src activation occurs before phosphorylation of EGFR. Blockade of Src activity by expression of dominant negative mutant of Src inhibits mitogen-activated protein kinase (MAPK) activation induced by TxA2. ERK activation induced by the PKC activator phorbol myristate acetate was inhibited by PTX, PP1, AG1478, and calphostin C. In contrast, activation of ERK by lysophosphatidic acid, a Gi-coupled receptor activator, was inhibited by PTX, PP1, and AG1478, but not by calphostin C. Thus, TP-stimulated ERK activation requires Gi, which in turn requires PKC activation. Immunoprecipitation of Galphai showed increased association of Galphai with TPalpha following PKC activation. In conclusion, TPalpha is directly coupled to the Gi protein by a PKC-regulated mechanism; Gi coupling causes Src-dependent transactivation of the EGFR, which is the dominant pathway in TP-mediated ERK activation.


Subject(s)
ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Genes, src/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Thromboxane/metabolism , Cell Line , ErbB Receptors/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genes, src/genetics , Humans , Indicators and Reagents , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/genetics , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/physiology , Tubulin/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
11.
Circ Res ; 87(9): 739-45, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11055976

ABSTRACT

Thromboxane A(2) (TxA(2)) causes platelet aggregation, vasoconstriction, and inhibition of endothelial cell (EC) migration and prevents vascular tube formation via its specific receptors (TP), of which there are two isoforms (TPalpha and TPbeta), both expressed in human ECs. In this study, we demonstrate that the TxA(2) mimetic IBOP increases apoptosis of human ECs and inhibits the phosphorylation of Akt kinase, an intracellular mediator required for cell survival. Treatment with IBOP destroyed EC networks formed on a basement membrane matrix in vitro. To distinguish the role of the TP isoforms, each isoform was expressed in TP-null ECs to create TPalpha and TPbeta ECs. IBOP induced apoptosis and inhibited phosphorylation of Akt kinase in both TPalpha and TPbeta. IBOP increased cAMP levels in TPalpha but not in TPbeta. Apoptosis induced by IBOP in TPalpha was not affected by either the adenylyl cyclase activator forskolin or the protein kinase A inhibitor 14-22 amide or H-89, whereas that in TPbeta was suppressed by forskolin and enhanced by the protein kinase A inhibitor 14-22 amide or H-89, suggesting that the TP isoforms differ in their signal pathways in mediating apoptosis. In conclusion, apoptosis may be the mechanism by which TxA(2)-mediated destruction of vascular structures in ECs occurs; although both TP isoforms induce apoptosis, possibly via inhibiting Akt phosphorylation, the signaling differs in each isoform, in that activation of the adenylyl cyclase pathway prevents apoptosis caused by TPbeta, but not by TPalpha, stimulation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis , Endothelium, Vascular/drug effects , Proto-Oncogene Proteins , Sulfonamides , Thromboxane A2/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Bridged Bicyclo Compounds, Heterocyclic , Cell Survival , Clone Cells , Colforsin/pharmacology , Cyclic AMP/metabolism , Endothelium, Vascular/cytology , Enzyme Activation , Fatty Acids, Unsaturated , Humans , Hydrazines/pharmacology , Isoquinolines/pharmacology , Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thromboxane A2/antagonists & inhibitors , Time Factors
13.
Adv Exp Med Biol ; 476: 327-50, 2000.
Article in English | MEDLINE | ID: mdl-10949677

ABSTRACT

The de-novo formation of vessels (angiogenesis) and the remodelling of preexisting collateral vessels (arteriogenesis) are processes that occur naturally in ischemic heart disease. Promoting these processes by administration of various substances or other physical stimuli (therapeutic angiogenesis) may provide a future strategy for the treatment of ischemic vascular diseases. Mechanisms of angiogenesis and arteriogenesis, as well as trials of therapeutic angiogenesis in animal models and humans are reviewed.


Subject(s)
Myocardial Ischemia/drug therapy , Neovascularization, Pathologic/physiopathology , Animals , Clinical Trials as Topic , Endothelial Growth Factors/metabolism , Fibroblast Growth Factors/metabolism , Humans , Lymphokines/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Biochem Biophys Res Commun ; 271(2): 499-508, 2000 May 10.
Article in English | MEDLINE | ID: mdl-10799325

ABSTRACT

Peptides based on the pseudosubstrate (PS) sequence of conventional protein kinase C isoenzymes (alpha, beta, gamma) specifically inhibit PKC activity in permeabilized cells, but whether PS can be used to study the role of PKC in the proliferation or migration of intact endothelial cells (EC) and angiogenesis is unknown. Peptides based on the PKCeta pseudosubstrate (etaPS) sequence were 3.5- to 8-fold more potent in inhibiting the PKCalpha, delta, epsilon, or eta kinase activity than was the peptide based on the PKCalpha pseudosubstrate (alphaPS) sequence. Thus, etaPS was conditionally overexpressed in intact EC and compared to alphaPS. Serum-induced growth of EC expressing etaPS was significantly slower than that of control EC. etaPS EC demonstrated slower rate of serum stimulated migration than that of either control or alphaPS EC. Expression of either etaPS or alphaPS produced slower rates of PMA induced EC migration, as compared to control EC. In an in vitro three-dimensional assay in which EC organize into capillary tubules, the EC that expressed etaPS formed fewer such tubules. This study shows that pseudosubstrate inhibitors derived from PKCeta are more potent both in vitro and in vivo than one based on the conventional isoenzyme PKCalpha. These data further support a role for PKC in proliferation and migration of intact EC, and angiogenesis.


Subject(s)
Endothelium, Vascular/cytology , Protein Kinase C/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Capillaries/enzymology , Cell Differentiation , Cell Division , Cell Line , Cell Movement , Cells, Cultured , Collagen/metabolism , Dexamethasone/pharmacology , Drug Combinations , Immunohistochemistry , Isoenzymes/metabolism , Kinetics , Laminin/metabolism , MAP Kinase Signaling System , Protein Kinase C/isolation & purification , Protein Kinase C/physiology , Protein Kinase C-alpha , Proteoglycans/metabolism , RNA/metabolism , Rats , Signal Transduction , Time Factors
16.
J Biol Chem ; 274(50): 35562-70, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10585431

ABSTRACT

The eicosanoid thromboxane A(2) (TXA(2)) is released by activated platelets, monocytes, and the vessel wall and interacts with high affinity receptors expressed in several tissues including endothelium. Whether TXA(2) might alter endothelial migration and tube formation, two determinants of angiogenesis, is unknown. Thus, we investigated the effect of the TXA(2) mimetic [1S-(1alpha, 2beta(5Z),3alpha(1E,3R), 4alpha]-7-[3-(3-hydroxy-4-(4'-iodophenoxy)-1-butenyl)-7-o xab icyclo- [2.2.1]heptan-2-yl]-5'-heptenoic acid (IBOP) on human endothelial cell (HEC) migration and angiogenesis in vitro. IBOP stimulation inhibited HEC migration by 50% and in vitro capillary formation by 75%. These effects of IBOP were time- and concentration-dependent with an IC(50) of 25 nM. IBOP did not affect integrin expression or cytoskeletal morphology of HEC. Since gap junction-mediated intercellular communication increases in migrating HEC, we determined whether IBOP might inhibit coupling or connexin expression in HEC. IBOP reduced the passage of microinjected dyes between HEC by 50%, and the effects of IBOP on migration and tube formation were mimicked by the gap junction inhibitor 18beta-glycyrrhetinic acid (1 microM) with a similar time course and efficacy. IBOP (24 h) did not affect the expression or phosphorylation of connexin 43 in whole HEC lysates. Immunohistologic examination of HEC suggested that IBOP may impair functional coupling by altering the cellular distribution of gap junctions, leading to increased connexin 43 internalization. Thus, this finding that TXA(2) mimetics can prevent HEC migration and tube formation, possibly by impairing intercellular communication, suggests that antagonizing TXA(2) signaling might enhance vascularization of ischemic tissue.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Capillaries/physiology , Cell Communication/drug effects , Chemotaxis/drug effects , Endothelium, Vascular/physiology , Fatty Acids, Unsaturated/pharmacology , Neovascularization, Physiologic/drug effects , Thromboxane A2/pharmacology , Calcium/metabolism , Capillaries/drug effects , Cells, Cultured , Connexin 43/metabolism , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gap Junctions/drug effects , Gap Junctions/physiology , Humans , Integrins/analysis , Umbilical Veins , Wound Healing/drug effects
17.
J Cell Biol ; 147(5): 1073-84, 1999 Nov 29.
Article in English | MEDLINE | ID: mdl-10579726

ABSTRACT

Migration, proliferation, and tube formation of endothelial cells are regulated by a protein kinase C isoenzyme PKCtheta. A full-length cDNA encoding a novel 20-kD protein, whose expression was PKCtheta-dependent, was identified in endothelial cells, cloned, characterized, and designated as theta-associated protein (TAP) 20. Overexpression of TAP20 decreased cell adhesion and enhanced migration on vitronectin and tube formation in three-dimensional culture. An antiintegrin alphavbeta5 antibody prevented these TAP20 effects. Overexpression of TAP20 also decreased focal adhesion formation in alphavbeta3-deficient cells. The interaction between TAP20 and beta5 integrin cytoplasmic domain was demonstrated by protein coprecipitation and immunoblotting. Thus, the discovery of TAP20, which interacts with integrin beta5 and modulates cell adhesion, migration, and tube formation, further defines a possible pathway to angiogenesis dependent on PKCtheta.


Subject(s)
Cell Movement/physiology , Integrin beta Chains , Integrins/physiology , Isoenzymes/physiology , Membrane Glycoproteins/physiology , Protein Kinase C/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion/physiology , Cell Division/physiology , Cloning, Molecular , Cytoplasm/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Integrins/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neovascularization, Physiologic/physiology , Nuclear Proteins , Protein Kinase C-theta , Proteins/chemistry , Rats , Receptors, Vitronectin/metabolism , Tumor Cells, Cultured , Zinc Fingers
18.
Circulation ; 100(18): 1865-71, 1999 Nov 02.
Article in English | MEDLINE | ID: mdl-10545430

ABSTRACT

BACKGROUND: Angiogenesis is a promising treatment strategy for patients who are not candidates for standard revascularization, because it promotes the growth of new blood vessels in ischemic myocardium. METHODS AND RESULTS: We conducted a randomized, double-blind, placebo-controlled study of basic fibroblast growth factor (bFGF; 10 or 100 microg versus placebo) delivered via sustained-release heparin-alginate microcapsules implanted in ischemic and viable but ungraftable myocardial territories in patients undergoing CABG. Twenty-four patients were randomized to 10 microg of bFGF (n=8), 100 microg of bFGF (n=8), or placebo (n=8), in addition to undergoing CABG. There were 2 operative deaths and 3 Q-wave myocardial infarctions. There were no treatment-related adverse events, and there was no rise in serum bFGF levels. Clinical follow-up was available for all patients (16.0+/-6.8 months). Three control patients had recurrent angina, 2 of whom required repeat revascularization. One patient in the 10-microg bFGF group had angina, whereas all patients in the 100-microg bFGF group remained angina-free. Stress nuclear perfusion imaging at baseline and 3 months after CABG showed a trend toward worsening of the defect size in the placebo group (20.7+/-3.7% to 23.8+/-5.7%, P=0.06), no significant change in the 10-microg bFGF group, and significant improvement in the 100-microg bFGF group (19.2+/-5.0% to 9.1+/-5.9%, P=0.01). Magnetic resonance assessment of the target ischemic zone in a subset of patients showed a trend toward a reduction in the target ischemic area in the 100-microg bFGF group (10.7+/-3.9% to 3. 7+/-6.3%, P=0.06). CONCLUSIONS: This study of bFGF in patients undergoing CABG demonstrates the safety and feasibility of this mode of therapy in patients with viable myocardium that cannot be adequately revascularized.


Subject(s)
Coronary Artery Bypass , Fibroblast Growth Factor 2/administration & dosage , Alginates , Coronary Vessels , Delayed-Action Preparations , Double-Blind Method , Drug Carriers , Drug Compounding , Drug Implants , Female , Follow-Up Studies , Glucuronic Acid , Heparin , Hexuronic Acids , Humans , Male , Middle Aged , Patient Selection , Placebos , Recombinant Proteins/administration & dosage
19.
Circulation ; 100(18): 1909-16, 1999 Nov 02.
Article in English | MEDLINE | ID: mdl-10545436

ABSTRACT

BACKGROUND: Although protein kinase C (PKC) has been implicated in ischemic cell death, the role of individual PKC isoenzymes in the response of endothelial cells (ECs) to hypoxia is unknown. METHODS AND RESULTS: To test the effect of hypoxia on the activity of individual PKC isoenzymes, human ECs were exposed to 95% N(2) with 5% CO(2) for 24 hours. This severe hypoxia reduced PKCdelta specific activity in both human umbilical vein ECs (HUVECs) and a HUVEC-derived EC line (ECVs) significantly (80.5+/-5.7% and 55.5+/-8. 6% of normoxia controls, respectively); the activities of PKCalpha and PKCepsilon were unchanged. The protein levels of PKCalpha, PKCdelta, and PKCepsilon were unchanged by hypoxia. To determine whether PKCdelta downregulation by hypoxia was linked to EC function, ECVs in which PKCdelta was stably overexpressed (PKCdelta-ECs) were exposed to hypoxia. A significant increase in cell death was observed in PKCdelta-ECs compared with controls (5.8+/-0.6% versus 2. 3+/-0.4% at 24 hours, 13.2+/-1.2% versus 4.1+/-0.4% at 48 hours, P<0. 05) during hypoxia. Neither the DNA laddering assay nor TUNEL staining revealed an increase in apoptosis of PKCdelta-ECs exposed to hypoxia, suggesting a hypoxia-induced increase in nonapoptotic cell death of PKCdelta-ECs. Inhibition of NO synthase with N(G)-monomethyl-L-arginine (L-NMMA) affected neither the decline in PKCdelta activity nor the EC death induced by hypoxia. CONCLUSIONS: PKCdelta activity is decreased by hypoxia by a mechanism that does not involve NO synthase; this downregulation appears to enhance EC survival during hypoxia by decreasing nonapoptotic cell death.


Subject(s)
Adaptation, Physiological , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Apoptosis/physiology , Cell Division/physiology , Down-Regulation , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Protein Kinase C-delta , omega-N-Methylarginine/pharmacology
20.
Circ Res ; 85(3): 247-56, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10436167

ABSTRACT

Vascular endothelial growth factor (VEGF) promotes angiogenesis and endothelial cell (EC) migration and proliferation by affecting intracellular mediators, only some of which are known, distal to its receptors. Protein kinase C (PKC) participates in the function of VEGF, but the role of individual PKC isoenzymes is unknown. In this study, we tested the importance of the activity of specific PKC isoenzymes in human EC migration and proliferation in response to VEGF. PKCdelta specific activity was depressed by the addition of VEGF (by 41+/-8% [P<0.05] at 24 hours) in human umbilical vein ECs (HUVECs) and in a HUVEC-derived EC line, ECV, without changing the total amount of either protein or mRNA encoding PKCdelta. Neither basic fibroblast growth factor (FGF-2) nor serum altered PKCdelta specific activity. The VEGF-induced decrease of PKCdelta activity, which began at 8 hours after stimulation, was strongly blocked by pretreatment with the nitric oxide (NO) synthase inhibitor N(G)-monomethyl-L-arginine in HUVECs; NO release peaked within 2 hours after stimulation. An exogenous NO donor, sodium nitroprusside, also decreased PKCdelta activity. The inhibition by N(G)-monomethyl-L-arginine of VEGF-induced HUVEC migration and proliferation, but not that induced by FGF-2 or serum, suggested that the decrease in PKCdelta via NO pathway is required for VEGF-induced EC migration and proliferation. Overexpression of PKCdelta in ECV cells specifically prevented EC response to VEGF but not to FGF-2 or serum. Thus, we conclude that suppression of PKCdelta activity via a NO synthase mechanism is required for VEGF-induced EC migration and proliferation, but not for that induced by FGF-2 or serum.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/physiology , Isoenzymes/antagonists & inhibitors , Lymphokines/pharmacology , Nitric Oxide/physiology , Protein Kinase C/antagonists & inhibitors , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroprusside/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-delta , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...