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










Publication year range
1.
J Clin Transl Sci ; 3(2-3): 65-74, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31544007

ABSTRACT

INTRODUCTION: We are developing the novel αIIbß3 antagonist, RUC-4, for subcutaneously (SC)-administered first-point-of-medical-contact treatment for ST Segment Elevated Myocardial Infarction (STEMI). METHODS: We studied the: 1. pharmacokinetics (PK) of RUC-4 at 1.0, 1.93, and 3.86 mg/kg IV, IM, and SC in non-human primates (NHPs); 2. impact of aspirin on RUC-4 IC50 in human platelet-rich plasma (PRP); 3. effect of different anticoagulants on the RUC-4 IC50 in human PRP; and 4. relationship between αIIbß3 receptor blockade by RUC-4 and inhibition of ADP-induced platelet aggregation. RESULTS: 1. All doses of RUC-4 were well tolerated, but animals demonstrated variable temporary bruising. IM and SC RUC-4 reached dose-dependent peak levels within 5-15 min, with T½ s between 0.28 and 0.56 hrs. Platelet aggregation studies in NHPs receiving IM RUC-4 demonstrated >80% inhibition of the initial slope of ADP-induced aggregation with all 3 doses 30 minutes post-dosing, with subsequent dose-dependent loss of inhibition over 4-5 hours. 2. The RUC-4 IC50 for ADP-induced platelet aggregation was unaffected by aspirin treatment (40±9 nM vs. 37±5 nM; p=0.39). 3. The RUC-4 IC50 was significantly higher in PRP prepared from PPACK-anticoagulated blood compared to citrate-anticoagulated blood using either TRAP (122±17 vs. 66±25 nM; p=0.05; n=4) or ADP (102±22 vs. 54±13; p<0.001; n=5). 4. There was a close correspondence between receptor blockade and inhibition of ADP-induced platelet aggregation, with aggregation inhibition beginning with ~40% receptor blockade and becoming nearly complete at >80% receptor blockade. DISCUSSION: Based on these results and others, RUC-4 has now progressed to formal preclinical toxicology studies.

2.
Front Pharmacol ; 8: 279, 2017.
Article in English | MEDLINE | ID: mdl-28659792

ABSTRACT

The 5-HT6 receptor is a promising target for cognitive disorders, in particular for Alzheimer's disease (AD) and other CNS disorders. The high-affinity and selective 5-HT6 receptor antagonist idalopirdine (Lu AE58054) is currently in development for mild-moderate AD as adjunct therapy to acetylcholinesterase inhibitors (AChEIs). We studied the effects of idalopirdine alone and in combination with the AChEI donepezil on brain activity using BOLD (Blood Oxygen Level Dependent) functional magnetic resonance imaging (fMRI) in the awake rat. Idalopirdine (2 mg/kg, i.v.) alone had a modest effect on brain activity, resulting in activation of eight brain regions at the peak response. Of these, the cholinergic diagonal band of Broca, the infralimbic cortex, the ventral pallidum, the nucleus accumbens shell, and the magnocellular preoptic area were shared with the effects of donepezil (0.3 mg/kg, i.v.). Donepezil alone activated 19 brain regions at the peak response, including several cortical regions, areas of the septo-hippocampal system and the serotonergic raphe nucleus. When idalopirdine and donepezil were combined, there was a robust stimulation pattern with activation of 36 brain regions spread across the extended-amygdala-, striato-pallidal, and septo-hippocampal networks as well as the cholinergic system. These findings indicate that, whilst idalopirdine and donepezil recruit a number of overlapping regions including one of the forebrain cholinergic nuclei, the synergistic effect of both compounds extends beyond the cholinergic system and the effects of donepezil alone toward recruitment of multiple neural circuits and neurotransmitter systems. These data provide new insight into the mechanisms via which idalopirdine might improve cognition in donepezil-treated AD patients.

3.
PLoS One ; 10(5): e0125748, 2015.
Article in English | MEDLINE | ID: mdl-25955025

ABSTRACT

Traumatic brain injury (TBI) can occur anywhere along the cortical mantel. While the cortical contusions may be random and disparate in their locations, the clinical outcomes are often similar and difficult to explain. Thus a question that arises is, do concussions at different sites on the cortex affect similar subcortical brain regions? To address this question we used a fluid percussion model to concuss the right caudal or rostral cortices in rats. Five days later, diffusion tensor MRI data were acquired for indices of anisotropy (IA) for use in a novel method of analysis to detect changes in gray matter microarchitecture. IA values from over 20,000 voxels were registered into a 3D segmented, annotated rat atlas covering 150 brain areas. Comparisons between left and right hemispheres revealed a small population of subcortical sites with altered IA values. Rostral and caudal concussions were of striking similarity in the impacted subcortical locations, particularly the central nucleus of the amygdala, laterodorsal thalamus, and hippocampal complex. Subsequent immunohistochemical analysis of these sites showed significant neuroinflammation. This study presents three significant findings that advance our understanding and evaluation of TBI: 1) the introduction of a new method to identify highly localized disturbances in discrete gray matter, subcortical brain nuclei without postmortem histology, 2) the use of this method to demonstrate that separate injuries to the rostral and caudal cortex produce the same subcortical, disturbances, and 3) the central nucleus of the amygdala, critical in the regulation of emotion, is vulnerable to concussion.


Subject(s)
Brain Concussion/pathology , Cerebral Cortex/injuries , Cerebral Cortex/pathology , Computer Simulation , Gray Matter/pathology , Imaging, Three-Dimensional , Amygdala/injuries , Amygdala/pathology , Animals , Anisotropy , Hippocampus/injuries , Hippocampus/pathology , Male , Percussion , Rats, Sprague-Dawley , Thalamus/injuries , Thalamus/pathology
4.
Front Syst Neurosci ; 9: 15, 2015.
Article in English | MEDLINE | ID: mdl-25745388

ABSTRACT

In the present study, we used functional MRI in awake rats to investigate the pain response that accompanies intradermal injection of capsaicin into the hindpaw. To this end, we used BOLD imaging together with a 3D segmented, annotated rat atlas and computational analysis to identify the integrated neural circuits involved in capsaicin-induced pain. The specificity of the pain response to capsaicin was tested in a transgenic model that contains a biallelic deletion of the gene encoding for the transient receptor potential cation channel subfamily V member 1 (TRPV1). Capsaicin is an exogenous ligand for the TRPV1 receptor, and in wild-type rats, activated the putative pain neural circuit. In addition, capsaicin-treated wild-type rats exhibited activation in brain regions comprising the Papez circuit and habenular system, systems that play important roles in the integration of emotional information, and learning and memory of aversive information, respectively. As expected, capsaicin administration to TRPV1-KO rats failed to elicit the robust BOLD activation pattern observed in wild-type controls. However, the intradermal injection of formalin elicited a significant activation of the putative pain pathway as represented by such areas as the anterior cingulate, somatosensory cortex, parabrachial nucleus, and periaqueductal gray. Notably, comparison of neural responses to capsaicin in wild-type vs. knock-out rats uncovered evidence that capsaicin may function in an antinociceptive capacity independent of TRPV1 signaling. Our data suggest that neuroimaging of pain in awake, conscious animals has the potential to inform the neurobiological basis of full and integrated perceptions of pain.

5.
Arterioscler Thromb Vasc Biol ; 34(10): 2321-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147334

ABSTRACT

OBJECTIVE: Treatment of myocardial infarction within the first 1 to 2 hours with a thrombolytic agent, percutaneous coronary intervention, or an αIIbß3 antagonist decreases mortality and the later development of heart failure. We previously reported on a novel small molecule αIIbß3 antagonist, RUC-2, that has a unique mechanism of action. We have now developed a more potent and more soluble congener of RUC-2, RUC-4, designed to be easily administered intramuscularly by autoinjector to facilitate its use in the prehospital setting. Here, we report the properties of RUC-4 and the antiplatelet and antithrombotic effects of RUC-2 and RUC-4 in animal models. APPROACH AND RESULTS: RUC-4 was ≈ 20% more potent than RUC-2 in inhibiting human ADP-induced platelet aggregation and much more soluble in aqueous solutions (60-80 mg/mL). It shared RUC-2's specificity for αIIbß3 versus αVß3, did not prime the receptor to bind fibrinogen, or induce changes in ß3 identified by a conformation-specific monoclonal antibody. Both RUC-2 and RUC-4 prevented FeCl3-induced thrombotic occlusion of the carotid artery in mice and decreased microvascular thrombi in response to laser injury produced by human platelets infused into transgenic mice containing a mutated von Willebrand factor that reacts with human but not mouse platelets. Intramuscular injection of RUC-4 in nonhuman primates at 1.9 and 3.85 mg/kg led to complete inhibition of platelet aggregation within 15 minutes, with dose-dependent return of platelet aggregation after 4.5 to 24 hours. CONCLUSIONS: RUC-4 has favorable biochemical, pharmacokinetic, pharmacodynamic, antithrombotic, and solubility properties as a prehospital therapy of myocardial infarction, but the possibility of increased bleeding with therapeutic doses remains to be evaluated.


Subject(s)
Blood Platelets/drug effects , Carotid Stenosis/prevention & control , Emergency Medical Services , Fibrinolytic Agents/pharmacology , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Pyrimidinones/pharmacology , Thiadiazoles/pharmacology , Thrombosis/prevention & control , Animals , Binding Sites , Blood Platelets/metabolism , Carotid Stenosis/blood , Carotid Stenosis/chemically induced , Chlorides , Disease Models, Animal , Ferric Compounds , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacokinetics , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Molecular Dynamics Simulation , Myocardial Infarction/blood , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Conformation , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Solubility , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Thiadiazoles/pharmacokinetics , Thrombosis/blood , Thrombosis/chemically induced , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
6.
Front Neurol ; 5: 94, 2014.
Article in English | MEDLINE | ID: mdl-25071696

ABSTRACT

Blood oxygen level dependent (BOLD) imaging in awake mice was used to identify differences in brain activity between wild-type, HETzQ175, and HOMzQ175 genotypes in response to the odor of almond. The study was designed to see how alterations in the huntingtin gene in a mouse model of Huntington's disease would affect the perception and processing of almond odor, an evolutionarily conserved stimulus with high emotional and motivational valence. Moreover, the mice in this study were "odor naïve," i.e., never having smelled almond or any nuts. Using a segmented, annotated MRI atlas of the mouse and computational analysis, 17 out of 116 brain regions were identified as responding differently to almond odor across genotypes. These regions included the glomerulus of the olfactory bulb, forebrain cortex, anterior cingulate, subiculum, and dentate gyrus of the hippocampus, and several areas of the hypothalamus. In many cases, these regions showed a gene-dose effect with HETzQ175 mice showing a reduction in brain activity from wild-type that is further reduced in HOMzQ175 mice. Conspicuously absent were any differences in brain activity in the caudate/putamen, thalamus, CA3, and CA1 of the hippocampus and much of the cortex. The glomerulus of the olfactory bulb in HOMzQ175 mice showed a reduced change in BOLD signal intensity in response to almond odor as compared to the other phenotypes suggesting a deficit in olfactory sensitivity.

7.
Bioorg Med Chem Lett ; 24(4): 1148-53, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24461295

ABSTRACT

A collection of αIIbß3 integrin receptor antagonists possessing a unique MIDAS metal ion displacement mechanism of action is presented. Insight into these agents' structure-activity relationships, binding modality, and pharmacokinetic and pharmacodynamic profiles highlight the potential of these small molecule ion displacement ligands as attractive candidates for clinical development.


Subject(s)
Blood Proteins/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Blood Proteins/chemical synthesis , Blood Proteins/chemistry , Dose-Response Relationship, Drug , Humans , Ions/chemistry , Ligands , Models, Molecular , Molecular Conformation , Platelet Aggregation/drug effects , Structure-Activity Relationship
8.
J Vasc Interv Radiol ; 15(2 Pt 1): 169-76, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14963184

ABSTRACT

PURPOSE: This study was designed to compare the ability of reteplase (a fibrinolytic agent) alone or in combination with abciximab (a monoclonal antibody antagonist of platelet glycoprotein IIb/IIIa) to achieve and sustain vessel patency in an acute model of peripheral arterial occlusive disease in cynomolgus monkeys. MATERIALS AND METHODS: Total arterial occlusion was induced in the femoral arteries of 32 cynomolgus monkeys (eight groups of four) by endothelial injury and injection of thrombin-treated autologous blood. Reteplase was administered by intravenous bolus dose or by intraarterial infusion at the site of the clot. Abciximab was administered as a single weight-adjusted intravenous bolus dose. Platelet activity was measured by ex vivo platelet aggregation before and after abciximab treatment. Different groups of animals received sequential partial doses of reteplase with or without increasing doses of abciximab until either the weight-adjusted human dose equivalent of reteplase was reached or vessel recanalization was achieved. RESULTS: Animals receiving reteplase-only regimens demonstrated variability in the times required for reperfusion and the permanence of the effect. The coadministration of abciximab at doses of the antibody that achieved near or full inhibition of platelet function generally decreased the time to reperfusion and resulted in more consistent and sustained vessel patency. In the case of systemic intravenous reteplase, the coadministration of abciximab resulted in effective reperfusion of thrombosed vessels at decreased doses of the lytic agent. CONCLUSIONS: Reteplase administered systemically or at the site of thrombotic occlusion restored blood flow for periods of varying duration in monkeys with acute femoral artery thrombosis. The coadministration of systemic intravenous abciximab to intravenous or intraarterial reteplase allowed the use of lower doses of fibrinolytic agent with more accelerated and sustained reperfusion.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arterial Occlusive Diseases/drug therapy , Fibrinolytic Agents/therapeutic use , Immunoglobulin Fab Fragments/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Recombinant Proteins/therapeutic use , Thrombosis/drug therapy , Tissue Plasminogen Activator/therapeutic use , Abciximab , Animals , Antibodies, Monoclonal/administration & dosage , Drug Therapy, Combination , Femoral Artery , Fibrinolytic Agents/administration & dosage , Immunoglobulin Fab Fragments/administration & dosage , Infusions, Intra-Arterial , Injections, Intravenous , Macaca fascicularis , Platelet Aggregation/drug effects , Recombinant Proteins/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Vascular Patency
9.
J Pharmacol Exp Ther ; 304(2): 855-61, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538843

ABSTRACT

Although it is well recognized that human platelet responses to alpha-thrombin are mediated by the protease-activated receptors PAR-1 and PAR-4, their role and relative importance in platelet-dependent human disease has not yet been elucidated. Because the expression profile of PARs in platelets from nonprimates differs from humans, we used cynomolgus monkeys to evaluate the role of PAR-1 in thrombosis. Based on reverse transcription-polymerase chain reaction, PAR expression in platelets from cynomolgus monkeys consisted primarily of PAR-1 and PAR-4, thereby mirroring the profile of human platelets. We probed the role of PAR-1 in a primate model of vascular injury-induced thrombosis with the selective PAR-1 antagonist (alpha S)-N-[(1S)-3-amino-1-[[(phenylmethyl)amino]carbonyl]propyl]-alpha-[[[[[1-(2,6-dichlorophenyl)methyl]-3-(1-pyrrolidinylmethyl)-1H-indazol-6-yl]amino]carbonyl]amino]-3,4-difluorobenzenepropanamide (RWJ-58259). After pretreatment with RWJ-58259 or vehicle, both carotid arteries of anesthetized monkeys were electrolytically injured and blood flow was monitored for 60 min. Time to occlusion was significantly extended after RWJ-58259 administration (27 +/- 3 to 53 +/- 8 min; p < 0.048). Vessels from three of the five treated animals remained patent. Ex vivo platelet aggregation measurements indicated complete PAR-1 inhibition, as well as an operational PAR-4 response. Immunohistochemical staining of mural thrombi with antibodies to the platelet marker CD61 and fibrinogen indicated that RWJ-58259 significantly reduced thrombus platelet deposition. Drug treatment had no effect on key hematological or coagulation parameters. Our results provide direct evidence that PAR-1 is the primary receptor that mediates alpha-thrombin's prothrombotic actions in primates and suggest that PAR-1 antagonists may have potential for the treatment of thrombotic disorders in humans.


Subject(s)
Carotid Artery, Common/drug effects , Receptors, Thrombin/antagonists & inhibitors , Thrombosis/prevention & control , Urea/analogs & derivatives , Animals , Carotid Artery, Common/pathology , Female , Indazoles/chemistry , Indazoles/pharmacology , Indazoles/therapeutic use , Macaca fascicularis , Male , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Receptor, PAR-1 , Thrombosis/drug therapy , Thrombosis/pathology , Urea/chemistry , Urea/pharmacology , Urea/therapeutic use
10.
J Am Coll Cardiol ; 40(2): 360-6, 2002 Jul 17.
Article in English | MEDLINE | ID: mdl-12106945

ABSTRACT

OBJECTIVES: This study was designed to assess the effect of abciximab on platelet and leukocyte deposition 60 min after stent insertion in nonhuman primates. BACKGROUND: Although it is well established that abciximab improves both short- and long-term clinical outcomes after stent placement, there have been no studies assessing its effect on early platelet and leukocyte deposition. METHODS: Cynomolgus monkeys were pretreated with aspirin and either saline or a 0.4 mg/kg bolus of abciximab, and then subjected to angioplasty and Palmaz-Schatz stent placement in the common iliac artery or abdominal aorta. After 60 min, animals were euthanized and the stented artery was evaluated by immunohistochemistry and morphometry. RESULTS: Complete occlusion of the stented vessel with a thin fibrin(ogen) meshwork and trapped blood occurred in two saline-treated and two abciximab-treated animals. In the four remaining saline-treated animals, a layer of erythrocytes trapped in a network of fibrin(ogen) was noted close to the vessel wall, and this was covered by a layer of large, irregular platelet thrombi. Leukocytes formed a monolayer on top of the platelets and near stent struts. In the four remaining abciximab-treated animals, the mean erythrocyte area was 65% smaller (p = 0.070), the platelet aggregate area was 89% smaller (p = 0.049) and the luminal area was 59% larger (p = 0.004). A monolayer of leukocytes also formed on top of the platelets and near stent struts. CONCLUSIONS: In control stented blood vessels in this study, platelet thrombi formed not at the vessel wall, but on top of an erythrocyte-rich layer, and platelets recruited leukocytes. Abciximab decreased the size of platelet thrombi, but did not prevent leukocyte recruitment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Anticoagulants/pharmacology , Arteries/pathology , Blood Platelets/drug effects , Immunoglobulin Fab Fragments/pharmacology , Leukocytes/drug effects , Platelet Aggregation Inhibitors/pharmacology , Stents/adverse effects , Thrombosis/prevention & control , Abciximab , Animals , Aorta, Abdominal/pathology , Arteries/drug effects , Arteries/injuries , Iliac Artery/pathology , Immunohistochemistry , Macaca fascicularis , Microscopy, Electron , Thrombosis/etiology , Time Factors
11.
Circ Res ; 90(4): 488-94, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11884380

ABSTRACT

A central role for leukocytes in neointimal hyperplasia after arterial injury is suspected. However, the relative importance of neutrophils and monocytes in balloon or stent-induced injury are not well understood, and mechanistic targeting of leukocyte recruitment or function is crude. We determined the temporal and spatial distribution of different leukocytes after balloon and stent-induced injury in primate iliac arteries. Based on these data, we targeted neutrophil and monocyte recruitment selectively after angioplasty or stent implantation and demonstrated that monocyte-specific blockade achieved via blockade of the MCP-1 receptor CCR2, was effective at reducing neointimal hyperplasia after stenting. In contrast, combined neutrophil and monocyte blockade achieved by targeting the leukocyte beta(2)-integrin beta-subunit CD18 was required to reduce neointimal hyperplasia after balloon injury. Distinct patterns of leukocyte infiltration in balloon versus stent-injured arteries predict distinct mechanisms for antiinflammatory strategies targeting neutrophils or monocytes in primates and may assist design of effective clinical strategies for optimizing vascular interventions.


Subject(s)
CD18 Antigens/drug effects , Graft Occlusion, Vascular/prevention & control , Leukocytes/metabolism , Receptors, Chemokine/antagonists & inhibitors , Stents , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blood Vessel Prosthesis Implantation , CD18 Antigens/biosynthesis , Catheterization/adverse effects , Disease Progression , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/pathology , Hyperplasia/etiology , Hyperplasia/pathology , Hyperplasia/prevention & control , Iliac Artery/pathology , Iliac Artery/surgery , Immunohistochemistry , Leukocytes/drug effects , Leukocytes/immunology , Macaca fascicularis , Macrophages/metabolism , Macrophages/pathology , Male , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Receptors, CCR2 , Receptors, Chemokine/biosynthesis , Stents/adverse effects , Tunica Intima/immunology , Tunica Intima/pathology , Tunica Intima/surgery , Vascular Patency/drug effects , Vascular Patency/immunology
12.
J Vasc Res ; 39(6): 524-33, 2002.
Article in English | MEDLINE | ID: mdl-12566978

ABSTRACT

Vascular access complications are a major problem in hemodialysis patients. Native arteriovenous fistulae, historically the preferred mode of access, have a patency rate of only 60% at 1 year. The most common mode of failure is due to progressive stenosis at the anastomotic site. We have previously demonstrated that perivascular endothelial cell implants inhibit intimal thickening following acute balloon injury in pigs and now seek to determine if these implants provide a similar benefit in the chronic and more complex injury model of arteriovenous anastomoses. Side-to-side femoral artery-femoral vein anastomoses were created in 24 domestic swine and the toxicological, biological and immunological responses to allogeneic endothelial cell implants were investigated 3 days and 1 and 2 months postoperatively. The anastomoses were wrapped with polymer matrices containing confluent porcine aortic endothelial cells (PAE; n = 14) or control matrices without cells (n = 10). PAE implants significantly reduced intimal hyperplasia at the anastomotic sites compared to controls by 68% (p < 0.05) at 2 months. The beneficial effects of the PAE implants were not due to differences in the rates of reendothelialization between the groups. No significant immunological response to the allogeneic endothelial cells that impacted on efficacy was detected in any of the pigs. No apparent toxicity was observed in any of the animals treated with endothelial implants. These data suggest that perivascular endothelial cell implants are safe and reduce early intimal hyperplasia in a porcine model of arteriovenous anastomoses.


Subject(s)
Arteriovenous Shunt, Surgical/methods , Endothelium, Vascular/transplantation , Femoral Artery/pathology , Femoral Artery/surgery , Animals , Antibodies/blood , Aorta/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Femoral Artery/immunology , Gelatin Sponge, Absorbable/pharmacology , Graft Survival/immunology , Hemostatics/pharmacology , Hyperplasia , T-Lymphocytes/immunology , Tunica Intima/immunology , Tunica Intima/pathology , Vasculitis/pathology
13.
J Thromb Thrombolysis ; 14(1): 15-24, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12652146

ABSTRACT

BACKGROUND: Tirofiban and eptifibatide are currently approved for the medical stabilization of non-ST segment elevation acute coronary syndromes. In patients undergoing percutaneous coronary intervention (PCI) during infusion of these drugs, conversion to abciximab, which has long term proven clinical efficacy and cost-effectiveness, following PCI may be desirable. The purpose of this study was to determine if the binding or pharmacodynamics of abciximab is affected by a prior infusion of either tirofiban or eptifibatide. METHODS: In vitro binding experiments were performed to determine if prior exposure to tirofiban or eptifibatide altered the affinity and extent of binding of abciximab to GPIIb/IIIa. For in vivo experiments, cynomolgus monkeys were pretreated with a bolus and 18 hour infusion of saline, tirofiban, or eptifibatide. At the end of the initial treatment, a bolus and 12 hr infusion of abciximab was started without delay. Inhibition of platelet aggregation, GPIIb/IIIa receptor blockade and abciximab pharmacokinetics were measured during and after both infusions. RESULTS: Equilibrium binding of abciximab in vitro was unaffected by tirofiban or eptifibatide. The extent and duration of abciximab inhibition of ex vivo platelet aggregation, receptor blockade, and abciximab pharmacokinetics in monkeys during and after the abciximab infusion were not affected by prior infusion of the animals with tirofiban or eptifibatide. CONCLUSIONS: In vitro and in vivo studies revealed that the molecular interaction of abciximab with the platelet GPIIb/IIIa receptor is not altered by immediate prior exposure of platelets to small molecule GPIIb/IIIa antagonists. These preclinical studies suggest that the efficacy of abciximab should not be impaired if it is initiated following termination of therapy with small molecule GPIIb/IIIa antagonists.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Abciximab , Animals , Binding Sites, Antibody/drug effects , Dose-Response Relationship, Drug , Drug Therapy, Combination , Eptifibatide , Female , Macaca fascicularis , Male , Peptides/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding/drug effects , Protein Binding/immunology , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...