Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
HIV AIDS (Auckl) ; 13: 789-800, 2021.
Article in English | MEDLINE | ID: mdl-34393518

ABSTRACT

BACKGROUND: In the past, protease inhibitors (PIs) and the reverse transcriptase inhibitor abacavir were identified increasing the risk for thromboembolic complications and cardiovascular events (CVE) of HIV infected patients taking a combination antiretroviral therapy (cART). Results of the previous HIV-PLA I-study lead to the assumption that platelet activation could play a substantial role in increasing CVE risks. METHODS: The open label, monocentric HIV-PLA II-study investigated HIV-1-infected, therapy-naïve adults (n=45) starting with cART, consisting either of boosted PI (atazanavir, n= 6, darunavir, n=11), NNRTI (efavirenz, n=14) or integrase inhibitor (raltegravir, n=14), each plus tenofovir/emtricitabine co-medication. Main exclusion criteria were tobacco smoking, the intake of NSAIDs or abacavir or past CVE. Platelet adhesive molecule p-selectin (CD62P) and FITC anti-human Integrin α-IIb/Integrin ß-3 (CD41/CD61) antibody (PAC-1) binding, monocyte CD11b/monocyte-associated CD41 expression and the endogenous thrombin potential (ETP) were assessed ex vivo-in vitro at baseline, weeks 4, 12 and 24. Therapy regimens were blinded to the investigators for laboratory and statistical analyses. RESULTS: CD11b and ETP showed no significant changes or differences between all study groups. In contrast, the mean + SD mean fluorescence units (MFI) of CD62P and PAC-1 increased significantly in patients taking PI, indicating an enhanced potential for thrombocyte activation and aggregation. CONCLUSION: CD62P expression, detecting the ɑ-platelet degranulation of pro-inflammatory and pro-thrombotic factors and adhesive proteins, and PAC-1 expression, representing a marker for conformation changes of the GIIb/IIIa receptor, increased significantly in patients taking HIV protease inhibitors. The findings of this study revealed a yet unknown pathway of platelet activation, possibly contributing to the increased risk for CVE under HIV protease inhibitor containing cART. CLINICAL TRIAL REGISTRATION NO: DRKS00000288.

2.
Eur J Clin Pharmacol ; 73(8): 949-956, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28523478

ABSTRACT

INTRODUCTION: Glycoprotein VI (GPVI) is the major platelet receptor for collagen-mediated platelet adhesion and activation. SAR264565 is an anti-GPVI-Fab, binds to GPVI with high affinity, and blocks GPVI function in human platelets in vitro. METHODS: The effect of SAR26456 on platelet responsiveness in the blood of 21 healthy male subjects was investigated using Sakariassen's ex vivo thrombogenesis perfusion chamber model on a collagen-coated surface under conditions mimicking arterial flow. Ex vivo effects of SAR264565 (10 and 100 µg/mL) were investigated before administration of aspirin or clopidogrel to study subjects (baseline), after aspirin (2× 300 mg) administration alone, and after combined aspirin (2× 300 mg)/clopidogrel (600 mg) administration. Additional ex vivo and in vitro platelet tests were also performed. RESULTS: Addition of SAR264565 to the perfusion chamber dose-dependently reduced platelet and fibrin deposition, reaching statistical significance at 100 µg/mL (415 ± 67 compared to 137 ± 36 platelets/cm2, [p < 0.01] and fibrin 0.095 ± 0.014 compared to 0.032 ± 0.008 µg/cm2, [p < 0.001]). Aspirin administration caused an additive and dose-dependent reduction of SAR264565-induced platelet and fibrin deposition. Combined aspirin/clopidogrel administration did not lead to additional SAR264565-induced inhibition of platelet or fibrin deposition. CONCLUSION: GPVI antagonism by the anti-GPVI-Fab fragment SAR264565 dose-dependently inhibits platelet adhesion and fibrin formation on a collagen surface under arterial shear. Additive inhibition is observed after prior aspirin administration with no further amplification on top of a combination of aspirin with clopidogrel. Ex vivo antiplatelet tests confirmed a selective inhibiting effect of SAR264565 on collagen-induced platelet activation.


Subject(s)
Blood Platelets/drug effects , Immunoglobulin Fab Fragments/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Adult , Blood Platelets/physiology , Fibrin/metabolism , Humans , Male , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/immunology , Young Adult
3.
Platelets ; 26(1): 59-66, 2015.
Article in English | MEDLINE | ID: mdl-24433287

ABSTRACT

The RGD cyclic pentapetide, cilengitide, is a selective inhibitor of αvß3 and αvß5 integrins and was developed for antiangiogenic therapy. Since cilengitide interacts with platelet αIIbß3 and platelets express αv integrins, the effect of cilengitide on platelet pro-coagulative response and adhesion is of interest. Flow-based adhesion assays were performed to evaluate platelet adhesion and rolling on von Willebrand factor (vWf), on fibrinogen and on human umbilical vein endothelial cells (HUVECs). Flow cytometry was used to detect platelet activation (PAC1) and secretion (CD62P) by cilengitide and light transmission aggregometry was used to detect cilengitide-dependent platelet aggregation. Cilengitide inhibited platelet adhesion to fibrinogen at concentrations above 250 µM [which is the Cmax in human studies] and adhesion to vWf and HUVECs at higher concentrations under physiologic flow conditions. Platelet aggregation was already impaired at cilengitide concentrations >10 µM. Activation of αIIbß3 integrin was inhibited by 250 µM cilengitide, whereas platelet secretion was unaffected by cilengitide. No evidence of cilengitide-induced platelet activation was found at all tested concentrations (0.01-1500 µM). At higher concentrations, platelet activation was inhibited, predominantly due to αIIbß3 inhibition.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Snake Venoms/pharmacology , Adenosine Diphosphate/pharmacology , Collagen/metabolism , Collagen/pharmacology , Endothelial Cells/metabolism , Fibrinogen/metabolism , Humans , P-Selectin/metabolism , Platelet Aggregation/drug effects , Receptors, Thrombin/metabolism
5.
ACS Med Chem Lett ; 3(9): 774-9, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-24900547

ABSTRACT

Exchange of the lipophilc part of ortho-substituted cinnamic acid lead structures with different small molecule fluorophoric moieties via a dimethylene spacer resulted in hEP3R ligands with affinities in the nanomolar concentration range. Synthesized compounds emit fluorescence in the blue, green, and red range of light and have been tested concerning their potential as a pharmacological tool. hEP3Rs were visualized by confocal laser scanning microscopy on HT-29 cells, on murine kidney tissues, and on human brain tissues and functionally were characterized as antagonists on human platelets. Inhibition of PGE2 and collagen-induced platelet aggregation was measured after preincubation with novel hEP3R ligands. The pyryllium-labeled ligand 8 has been shown as one of the most promising structures, displaying a useful fluorescence and highly affine hEP3R antagonists.

6.
Blood ; 117(18): 4999-5008, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21398580

ABSTRACT

The membrane-anchored CX3C chemokine fractalkine (FKN) is expressed on activated endothelium and is associated with the development of atherosclerosis. The potential of FKN in mediating platelet adhesion beyond platelet activation remains unexplored to date. A flow-based adhesion assay was used to study the adhesion of platelets to immobilized FKN under physiologic flow conditions. Platelet adhesion to von Willebrand factor (VWF) was increased in the presence of FKN at 600 inverse seconds. Additional platelet adhesion to FKN coimmobilized with VWF was dependent on the FKN receptor CX3CR1 and activation of glycoprotein (GP) IIb/IIIa. The number of platelets rolling on VWF was likewise enhanced in the presence of FKN. The enhancement of rolling on FKN and VWF was insensitive to anti-CX3CR1 antibody but was fully inhibited by neutralizing GPIbα function. The extracellular domain of GPIbα was covalently coupled to fluorescent microspheres, and microsphere binding was significantly higher in the presence of FKN. Platelet adhesion to activated endothelium in vitro and to intact human arteries was substantially increased in an FKN-dependent manner. These data demonstrate that endothelial expressed FKN activates platelets via its cognate receptor CX3CR1, whereas platelet adhesion is predominantly mediated by GPIbα and independent of CX3CR1.


Subject(s)
Chemokine CX3CL1/physiology , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins/physiology , von Willebrand Factor/physiology , Arteries/physiology , CX3C Chemokine Receptor 1 , Endothelial Cells/physiology , Hemorheology , Humans , In Vitro Techniques , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/physiology , Receptors, HIV/antagonists & inhibitors , Receptors, HIV/physiology
7.
Thromb Res ; 127(2): 105-10, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21144558

ABSTRACT

P-selectin glycoprotein ligand-1 (PSGL-1) interacts with P-selectin expressed on endothelial cells and platelets. PSGL-1 extracellular mucin-like domain displays a variable number of tandem repeats (VNTRs) polymorphism. The wildtype consists of 16 decameric repeats (designated A isoforms) and variants with 15 (B allele) and 14 (C allele) repeats that are assumed to be associated with reduced risk of vascular disease. We investigated the adhesion of these natural variants to P-selectin in native human neutrophils. Healthy volunteers were genotyped and the adhesion of neutrophils expressing the PSGL-1 isoforms A/A, A/B and B/B were studied under static and physiologic flow conditions. Homozygous B/B neutrophils attached significantly weaker to P-selectin at elevated shear rates from 24 up to 64 dyn/cm(2) than A/A and A/B neutrophils. No difference in adhesion rate was found under static conditions and shear stress below 24 dyn/cm(2), but B/B neutrophils rolled significantly faster than A/A neutrophils at shear stress ≥ 12 dyn/cm(2). There was no difference in the adhesive capacity between A/A an A/B neutrophils. These data support the view that the role of the decamers is to extend the ligand binding domain far above the cell surface to support stable leukocyte adhesion and rolling.


Subject(s)
Alleles , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Neutrophils/physiology , P-Selectin/blood , Adult , Amino Acid Sequence , Animals , CHO Cells , Cell Adhesion/physiology , Cricetinae , Cricetulus , Female , Flow Cytometry , Humans , Male , Minisatellite Repeats , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/metabolism , Polymorphism, Genetic , Protein Isoforms , Young Adult
8.
Pain ; 147(1-3): 116-21, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19762153

ABSTRACT

We investigated the analgesic efficacy of single doses of ibuprofen, tramadol and pregabalin in menthol-evoked cold pain in a randomized, placebo-controlled four-way cross-over study in 20 healthy volunteers. Tramadol 100mg significantly reduced menthol-evoked cold hyperalgesia. Effects of ibuprofen 600mg and pregabalin 100mg were not significant. Analgesic effects of tramadol were associated with minor side effects, particularly fatigue and nausea. Minor side effects also accompanied analgesic effects of pregabalin and ibuprofen in subjects responding to these drugs, mostly fatigue, dizziness and difficulties to concentrate for pregabalin and gastric upset for ibuprofen. Five out of 18 subjects had a 50% reduction of cold hyperalgesia with tramadol, three of these additionally responded to pregabalin, and two with all three drugs. The numbers needed to treat (NNT >or= 50% for tramadol 4.5, for pregabalin 9) largely agree with the reported efficacy of tramadol and of moderate dosages of pregabalin in patients with peripheral or central neuropathic pain suggesting that menthol-evoked cold pain hypersensitivity may represent a valid model for neuropathic pain, particularly cold allodynia.


Subject(s)
Analgesics/therapeutic use , Cold Temperature/adverse effects , Hyperalgesia , Ibuprofen/therapeutic use , Menthol , Tramadol/therapeutic use , gamma-Aminobutyric Acid/analogs & derivatives , Adult , Analysis of Variance , Area Under Curve , Case-Control Studies , Cross-Over Studies , Double-Blind Method , Female , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/psychology , Male , Pain Measurement/methods , Pain Threshold/drug effects , Physical Stimulation/methods , Pregabalin , Time Factors , gamma-Aminobutyric Acid/therapeutic use
9.
J Clin Pharmacol ; 48(10): 1189-96, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18716314

ABSTRACT

Low-molecular-weight heparins (LMWHs) differ considerably in their influence on clotting tests and release of tissue factor pathway inhibitor (TFPI). Biosimilarity therefore becomes an issue when generic forms of LMWHs are developed. So far, no bioequivalence study with a generic LMWH has been reported. A generic enoxaparin (test) was compared with the originator (reference) in 20 volunteers after single-dose subcutaneous administration (40 mg enoxaparin sodium, 4000 IU/mL anti-factor Xa (anti-FXa; activity). Target variables were anti-FXa and anti-FIIa activity, activated partial thromboplastin time (aPTT), prothrombinase-induced clotting time (PiCT), and TFPI over 24 hours. The statistical evaluation of the anti-FXa activity profile demonstrated bioequivalence of test and reference with confidence intervals of area under the plasma concentration-time curve (AUC0-tlast) (93%-99%) and Amax (88%-95%). Confidence intervals of AUC(0-tlast) (89%-102%) and Amax (90%-103%) of anti-FIIa activity also fulfill bioequivalence criteria. The 90% confidence interval for the maximum concentration of TFPI ranged from 90% to 113%. The claim of similarity was also supported by aPTT and PiCT profiles. Bioequivalence with the originator enoxaparin could be demonstrated by ex vivo inhibition of FXa and FIIa activity, by coagulation tests (aPTT and PiCT), and by in vivo release of TFPI. Whether such data also prove biosimilarity of the generic enoxaparin needs to be determined.


Subject(s)
Anticoagulants/pharmacokinetics , Blood Coagulation/drug effects , Drugs, Generic/pharmacokinetics , Enoxaparin/pharmacokinetics , Adult , Anticoagulants/administration & dosage , Anticoagulants/pharmacology , Area Under Curve , Blood Coagulation Tests/methods , Cross-Over Studies , Double-Blind Method , Drugs, Generic/administration & dosage , Drugs, Generic/pharmacology , Enoxaparin/administration & dosage , Enoxaparin/pharmacology , Factor Xa Inhibitors , Female , Humans , Lipoproteins/drug effects , Male , Partial Thromboplastin Time , Prothrombin/antagonists & inhibitors , Therapeutic Equivalency
10.
J Antimicrob Chemother ; 62(5): 1118-21, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18753189

ABSTRACT

INTRODUCTION: Thromboembolic complications under antiretroviral therapy (ART) have been described in the past. In particular, the influence of protease inhibitors (PIs) on platelet activation and coagulation is currently under discussion. METHODS: HIV-1-infected, PI-naive adults (n = 18) were investigated before and 4 weeks after the start of the ART, consisting either of boosted PI regimens (n = 13) plus reverse transcriptase inhibitors (RTIs) or a double PI regimen (n = 5) without RTI co-medication. Administered PIs were saquinavir (n = 15), lopinavir (n = 4), fosamprenavir (n = 2) and atazanavir (n = 2). Platelet CD62P, CD40L (%+ cells) and PAC-1 binding [mean fluorescence intensity (MFI)] as well as monocyte CD11b (MFI) and monocyte-associated CD41 (%+ cells and MFI) expression were assessed by flow cytometry with or without platelet stimulation. To investigate the influence of platelets on coagulation, the endogenous thrombin potential (ETP) [render fluorescence units (RFI)] was determined. RESULTS: CD62P, PAC-1 binding and CD11b expression remained unchanged. In contrast, the mean+/-SD MFI of CD40L (from 18.2+/-9.0 to 25.5+/-10.4, P = 0.038) and CD41 (from 446.1+/-213.8 to 605.0+/-183.8, P = 0.010) as markers for increased platelet-leucocyte interaction increased significantly. The collagen-induced ETP time-to-peak was altered significantly from 23.8+/-11.4 to 17.0+/-4.2 min (P = 0.028), although the ETP RFI peak showed no evidence for increased procoagulatory capacity (47.1+/-18.6 to 57.3+/-19.9, P = 0.085). CONCLUSIONS: Effects of the evaluated PI HIV therapy on platelet function assessed under field conditions seem to be minor, not affecting all investigated parameters. We found no evidence for increased platelet activation under PI-containing ART. However, CD41 as a marker for increased platelet-leucocyte interaction and CD40L, which can contribute to atherosclerosis, increased significantly.


Subject(s)
Blood Platelets/drug effects , Cell Adhesion Molecules/biosynthesis , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Leukocytes/drug effects , Adult , Blood Platelets/chemistry , CD11b Antigen/analysis , CD40 Ligand/analysis , Female , Flow Cytometry , Humans , Leukocytes/chemistry , Male , Middle Aged , P-Selectin/analysis , Platelet Membrane Glycoprotein IIb/analysis
11.
J Antimicrob Chemother ; 61(2): 394-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18156609

ABSTRACT

OBJECTIVES: In the past, bleeding events have been described for patients with haemophilia taking HIV-1 protease inhibitors. Recently, the FDA published a warning concerning intracranial haemorrhage in patients taking the HIV-1 protease inhibitor tipranavir co-administered with ritonavir. METHODS: We investigated (i) platelet aggregation in vivo in HIV-1-infected adult patients (n = 5) immediately before and 2 and 4 h after dosing of tipranavir/ritonavir 500/200 mg. To further characterize the effects, we then evaluated (ii) platelet aggregation and (iii) thromboxane B2 (TxB2) formation (ELISA) with increasing tipranavir concentrations (TPV(conc)) in vitro of up to 100,000 ng/mL. Platelet aggregation was stimulated either with 2 microM ADP (ADP) or 10 mg/L collagen (COL). TPV(conc) were measured with validated EPI-LC-MS/MS. Intraindividual comparisons of values at time points and TPV(conc), respectively, were carried out with repeated samples ANOVA. RESULTS: Platelet aggregation (mean, maximal light transmission A(max)) was significantly decreased in patients 4 h post-dose in collagen- (from 79.8% to 57.1%; P < 0.001) and in ADP-stimulated (from 58.5% to 54.0%; not significant) samples at a median (range) TPV(conc) of 62 500 ng/mL (22,990-67,500). These results could be reproduced in vitro at TPV(conc) 50,000 ng/mL (A(max)ADP/A(max)COL = 20.7/36.9%; P = 0.003/<0.001) and 100 000 ng/mL (A(max)ADP/A(max)COL = 14.5/17.1%; P < 0.001/<0.001). Median (range) TxB2 concentrations were reduced (P = 0.07) from 327 ng/mL (187-500) at baseline to 265 ng/mL (152-428) at 5000 ng/mL and were significantly reduced (P < 0.001) to 187 ng/mL (81-362) at a TPV(conc) of 50,000 ng/mL, respectively. CONCLUSIONS: Five HIV-1-infected patients on tipranavir-containing highly active antiretroviral therapy presented marked decreases in platelet aggregation. In vitro these effects were reproduced and decreased TxB2 formation was also demonstrated. Inhibition of platelet aggregation while receiving tipranavir treatment might contribute to increased risk of bleeding.


Subject(s)
Platelet Aggregation/drug effects , Pyridines/pharmacology , Pyrones/pharmacology , Thromboxane B2/blood , Adult , HIV Infections/blood , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , HIV-1 , Humans , Male , Middle Aged , Platelet Aggregation/physiology , Pyridines/therapeutic use , Pyrones/therapeutic use , Sulfonamides
12.
Thromb Res ; 111(4-5): 251-7, 2003.
Article in English | MEDLINE | ID: mdl-14693172

ABSTRACT

The sodium-hydrogen exchanger isoform 1 (NHE-1) contributes to platelet activation at elevated pH. Effects of NHE-1 inhibitors on platelet degranulation and formation of proinflammatory and procoagulatory platelet-leukocyte aggregates (PLA) and possible interactions with P2Y(12) inhibitors--which also affect platelet degranulation--have not been investigated. Whole blood from healthy human subjects was incubated with the NHE-1 inhibitor cariporide and the P2Y(12) inhibitor AR-C 69331 MX at clinically reasonable concentrations, in the presence of normal pH or in a propionate model to activate the NHE-1 (approximately pH 7.0). The degranulation marker CD62p, the expression of the activated GPIIb/IIIa receptor (PAC-1), and formation of platelet-leukocyte (monocyte) aggregates (PLA) was assessed by flow cytometry. Cariporide at concentrations up to 20 microg/ml had no effects on ADP- (5 microM) or TRAP- (2 microM) induced CD62p expression or PLA formation at normal pH. At pH 7.0 and stimulation with ADP, PLA decreased from 64+/-24% (control) to 47+/-23% under cariporide at 2 microg/ml (p<0.05), and the MFI of PLA (i.e. the platelet mass attached at monocytes) decreased from 547+/-203 to 360+/-96 units (p<0.05). PAC-1 MFI decreased from 66+/-23 to 34+/-18 units (p<0.05) after ADP and from 74+/-29 to 42+/-17 units (p<0.05) after TRAP, respectively. AR-C 69331 MX (10 nM) had inhibitory effects on all parameters irrespectively of the pH, and the combination of both agents at pH 7.0 shows additive effects. In conclusion, our investigation points to-perhaps clinically relevant-effects of NHE-1 inhibition on the degranulation of platelets and formation of platelet-leukocyte aggregates.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Blood Platelets/metabolism , Guanidines/pharmacology , Leukocytes/metabolism , P-Selectin/blood , Platelet Aggregation/drug effects , Receptors, Pituitary Hormone/blood , Sulfones/pharmacology , Blood Platelets/drug effects , Cell Aggregation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Combinations , Humans , Leukocytes/drug effects , Membrane Proteins/antagonists & inhibitors , Neutrophil Activation/drug effects , Osmosis/drug effects , P-Selectin/metabolism , Purinergic P2 Receptor Antagonists , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/metabolism , Receptors, Purinergic P2Y12 , Sodium-Hydrogen Exchangers/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...