Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Johann Bartko is active.

Publication


Featured researches published by Johann Bartko.


Thrombosis and Haemostasis | 2011

Increased platelet aggregation and in vivo platelet activation after granulocyte colony-stimulating factor administration. A randomised controlled trial.

Alexander O. Spiel; Johann Bartko; Michael Schwameis; Christa Firbas; Jolanta M. Siller-Matula; Matthias Schuetz; Manuela Weigl; Bernd Jilma

Granulocyte colony-stimulating factor (G-CSF) stimulates the bone marrow to produce granulocytes and stem cells and is widely used to accelerate neutrophil recovery after chemotherapy. Interestingly, specific G-CSF receptors have been demonstrated not only on myeloid cells, but also on platelets. Data on the effects of G-CSF on platelet function are limited and partly conflicting. The objective of this study was to determine the effect of G-CSF on platelet aggregation and in vivo platelet activation. Seventy-eight, healthy volunteers were enrolled into this randomised, placebo-controlled trial. Subjects received 5 μg/kg methionyl human granulocyte colony-stimulating factor (r-metHuG-CSF, filgrastim) or placebo subcutaneously for four days. We determined platelet aggregation with a whole blood impedance aggregometer with various, clinically relevant platelet agonists (adenosine diphosphate [ADP], collagen, arachidonic acid [AA], ristocetin and thrombin receptor activating peptide 6 [TRAP]). Filgrastim injection significantly enhanced ADP (+40%), collagen (+60%) and AA (+75%)-induced platelet aggregation (all p<0.01 as compared to placebo and p<0.001 as compared to baseline). In addition, G-CSF enhanced ristocetin-induced platelet aggregation (+18%) whereas TRAP-induced platelet aggregation decreased slightly (-14%) in response to filgrastim. While baseline aggregation with all agonists was only slightly but insignificantly higher in women than in men, this sex difference was enhanced by G-CSF treatment, and became most pronounced for ADP after five days (p<0.001). Enhanced platelet aggregation translated into a 75% increase in platelet activation as measured by circulating soluble P-selectin. G-CSF enhances platelet aggregation and activation in humans. This may put patients suffering from cardiovascular disease and cancer at risk for thrombotic events.


Journal of Cardiovascular Magnetic Resonance | 2011

Effect of ischemic preconditioning in skeletal muscle measured by functional magnetic resonance imaging and spectroscopy: a randomized crossover trial

Martin Andreas; Albrecht Ingo Schmid; M Keilani; Daniel Doberer; Johann Bartko; Richard Crevenna; Ewald Moser; Michael Wolzt

BackgroundNuclear magnetic resonance (NMR) imaging and spectroscopy have been applied to assess skeletal muscle oxidative metabolism. Therefore, in-vivo NMR may enable the characterization of ischemia-reperfusion injury. The goal of this study was to evaluate whether NMR could detect the effects of ischemic preconditioning (IPC) in healthy subjects.MethodsTwenty-three participants were included in two randomized crossover protocols in which the effects of IPC were measured by NMR and muscle force assessments. Leg ischemia was administered for 20 minutes with or without a subsequent impaired reperfusion for 5 minutes (stenosis model). IPC was administered 4 or 48 hours prior to ischemia. Changes in 31phosphate NMR spectroscopy and blood oxygen level-dependent (BOLD) signals were recorded. 3-Tesla NMR data were compared to those obtained for isometric muscular strength.ResultsThe phosphocreatine (PCr) signal decreased robustly during ischemia and recovered rapidly during reperfusion. In contrast to PCr, the recovery of muscular strength was slow. During post-ischemic stenosis, PCr increased only slightly. The BOLD signal intensity decreased during ischemia, ischemic exercise and post-ischemic stenosis but increased during hyperemic reperfusion. IPC 4 hours prior to ischemia significantly increased the maximal PCr reperfusion signal and mitigated the peak BOLD signal during reperfusion.ConclusionsIschemic preconditioning positively influenced muscle metabolism during reperfusion; this resulted in an increase in PCr production and higher oxygen consumption, thereby mitigating the peak BOLD signal. In addition, an impairment of energy replenishment during the low-flow reperfusion was detected in this model. Thus, functional NMR is capable of characterizing changes in reperfusion and in therapeutic interventions in vivo.Trial RegistrationClinicalTrials.gov: NCT00883467


Clinical Science | 2012

Effects of prasugrel on platelet inhibition during systemic endotoxaemia: a randomized controlled trial

Alexander O. Spiel; Ulla Derhaschnig; Michael Schwameis; Johann Bartko; Jolanta M. Siller-Matula; Bernd Jilma

P2Y(12) receptor antagonists have become a mainstay for the treatment of CVD (cardiovascular diseases). However, they have rarely been evaluated under pathophysiological conditions apart from arterial diseases. We hypothesized interactions between prasugrel and enhanced vWF (von Willebrand Factor) release in a model of systemic inflammation, and compared the pharmacodynamic effects of prasugrel against placebo on agonist-induced platelet aggregation and shear-induced platelet plug formation. A total of 20 healthy male volunteers were enrolled in a double-blind placebo-controlled two-way crossover trial. Each volunteer received either placebo or a 60 mg loading dose of prasugrel 2 h before endotoxin or placebo infusion. Platelet inhibition was measured with MEA (multiple electrode aggregometry), the PFA-100 system and the VASP (vasodilator-stimulated phosphoprotein) phosphorylation assay. Prasugrel blunted various platelet aggregation pathways, including those induced by ADP (-81%), AA (arachidonic acid) (-60%), ristocetin (-75%; P<0.001 for all) and, to a lesser degree, collagen or TRAP (thrombin-receptor-activating peptide). Prasugrel decreased shear-induced platelet plug formation, but vWF release during endotoxaemia partly antagonized the inhibitory effect of prasugrel as measured with the PFA-100 system. Endotoxaemia acutely decreased ristocetin and TRAP-induced platelet aggregation, and enhanced ristocetin-induced aggregation after 24 h. Strong in vivo blockade of P2Y(12) inhibits a broad spectrum of platelet aggregation pathways. However, vWF release may reduce prasugrels effects under high-shear conditions.


Transfusion | 2013

Recovery, safety, and tolerability of a solvent/detergent‐treated and prion‐safeguarded transfusion plasma in a randomized, crossover, clinical trial in healthy volunteers

Petra Jilma-Stohlawetz; Friedrich W. Kursten; Michaela Horvath; Gerda Leitner; Jana List; Jana Marcek; Peter Quehenberger; Michael Schwameis; Johann Bartko; Ulla Derhaschnig; Bernd Jilma

Octaplas LG is a prion‐depleted version of a previous generation product called Octaplas S/D. We compared the recovery, safety, and tolerability of these two pharmaceutical‐grade plasmas.


Transfusion | 2011

Safety of a universal, virus-inactivated and prion-depleted, pharmaceutical-quality plasma: a randomized, double-blind, clinical trial in healthy volunteers.

Petra Jilma-Stohlawetz; Friedrich W. Kursten; Claudia Walasek; Michaela Horvath; Gerda Leitner; Jana List; Peter Quehenberger; Michael Schwameis; Johann Bartko; Bernd Jilma

BACKGROUND: Universal plasma is intended to be transfused irrespective of the blood group. We compared the safety and tolerability of a novel, universal blood group–independent plasma with an ABO‐matched plasma.


British Journal of Clinical Pharmacology | 2016

Dissociation between systemic and pulmonary anti‐inflammatory effects of dexamethasone in humans

Johann Bartko; Leopold Stiebellehner; Ulla Derhaschnig; Christian Schoergenhofer; Michael Schwameis; Helmut Prosch; Bernd Jilma

Aims The local pulmonary inflammatory response has a different temporal and qualitative profile compared with the systemic inflammatory response. Although glucocorticoids substantially downregulate the systemic release of acute‐phase mediators, it is not clear whether they have comparable inhibitory effects in the human lung compartment. Therefore, we compared the anti‐inflammatory effects of a pure glucocorticoid agonist, dexamethasone, on bronchoalveolar lavage and blood cytokine concentrations in response to bronchially instilled endotoxin. Methods In this randomized, double‐blind and placebo‐controlled trial, 24 volunteers received dexamethasone or placebo and had endotoxin instilled into a lung segment and saline instilled into a contralateral segment, followed by bronchoalveolar lavage. Results Bronchially instilled endotoxin induced a local and systemic inflammatory response. Dexamethasone strongly blunted the systemic interleukin (IL) 6 and C‐reactive protein release. In sharp contrast, dexamethasone left the local release of acute‐phase mediators in the lungs virtually unchanged: bronchoalveolar lavage levels of IL‐6 were only 18% lower and levels of IL‐8 were even higher with dexamethasone compared with placebo, although the differences between treatments were not statistically significant (P = 0.07 and P = 0.08, respectively). However, dexamethasone had inhibitory effects on pulmonary protein extravasation and neutrophil migration. Conclusions The present study demonstrated a remarkable dissociation between the systemic anti‐inflammatory effects of glucocorticoids and its protective effects on capillary leak on the one hand and surprisingly low anti‐inflammatory effects in the lungs on the other.


Transplantation | 2017

Blockade of HLA Antibody-Triggered Classical Complement Activation in Sera From Subjects Dosed With the Anti-C1s Monoclonal Antibody TNT009—Results from a Randomized First-in-Human Phase 1 Trial

Jakob Mühlbacher; Bernd Jilma; Markus Wahrmann; Johann Bartko; Farsad Eskandary; Christian Schörgenhofer; Michael Schwameis; Graham C. Parry; James C. Gilbert; Sandip Panicker; Georg A. Böhmig

Background Complement may play a key role in antibody-mediated rejection. A promising therapeutic approach may be classical pathway (CP) inhibition at the level of early component C1. Methods In this first-in-human, double-blind, randomized placebo-controlled phase 1 trial, we evaluated the safety and complement inhibitory effect of TNT009, a humanized monoclonal anti-C1s antibody. Sixty-four adult healthy volunteers received either single (n = 48; 7 consecutive cohorts, 0.3-100 mg/kg) or 4 weekly infusions (n = 16; 2 consecutive cohorts, 30 and 60 mg/kg per infusion) of TNT009 or placebo. To assess the effect of treatment on complement activity, sera from dosed subjects were analyzed in a CP activation assay evaluating C3d deposition on HLA-coated microbeads spiked with alloantibodies. Results Single doses of TNT009 at 3 to 100 mg/kg uniformly and profoundly inhibited HLA antibody-mediated C3d deposition (≥86% after 60 minutes), whereby the duration of CP inhibition (2-14 days) was dose-dependent. Four weekly doses persistently blocked complement for 5 to 6 weeks. Ex vivo serum CP activity was profoundly inhibited when TNT009 concentrations exceeded 20 &mgr;g/mL. Infusions were well tolerated without serious or severe adverse events. Conclusions Treatment with TNT009 was safe and potently inhibited CP activity. Future studies in patients are required to assess the potential of TNT009 for preventing or treating antibody-mediated rejection.BACKGROUND Complement may play a key role in antibody-mediated rejection (ABMR). A promising therapeutic approach may be classical pathway (CP) inhibition at the level of early component C1. METHODS In this first-in-human, double-blind, randomized placebo-controlled phase 1 trial, we evaluated the safety and complement inhibitory effect of TNT009, a humanized monoclonal anti-C1s antibody. Sixty-four adult healthy volunteers received either single (n=48; 7 consecutive cohorts: 0.3-100 mg/kg) or 4 weekly infusions (n=16; 2 consecutive cohorts: 30 and 60 mg/kg per infusion) of TNT009 or placebo. To assess the effect of treatment on complement activity, sera from dosed subjects were analysed in a CP activation assay evaluating C3d deposition on HLA-coated microbeads spiked with alloantibodies. RESULTS Single doses of TNT009 at 3-100 mg/kg uniformly and profoundly inhibited HLA antibody-mediated C3d deposition (≥86% after 60 min), whereby the duration of CP inhibition (2-14 days) was dose-dependent. Four weekly doses persistently blocked complement for 5-6 weeks. Ex vivo serum CP activity was profoundly inhibited when TNT009 concentrations exceeded 20 μg/mL. Infusions were well tolerated without serious or severe adverse events. CONCLUSIONS Treatment with TNT009 was safe and potently inhibited CP activity. Future studies in patients are required to assess the potential of TNT009 for preventing or treating ABMR.This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.


Clinical Pharmacology & Therapeutics | 2018

A Randomized, First‐in‐Human, Healthy Volunteer Trial of sutimlimab, a Humanized Antibody for the Specific Inhibition of the Classical Complement Pathway

Johann Bartko; Christian Schoergenhofer; Michael Schwameis; Christa Firbas; Martin Beliveau; Colin Chang; Jean-Francois Marier; Darrell Nix; James C. Gilbert; Sandip Panicker; Bernd Jilma

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody‐mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement‐mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first‐in‐human, double‐blind, randomized, placebo‐controlled, dose‐escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration–effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 μg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Scandinavian Journal of Clinical & Laboratory Investigation | 2017

Evaluation of between-, within- and day-to-day variation of coagulation measured by rotational thrombelastometry (ROTEM)

Petra Jilma-Stohlawetz; Susanne Fritsche-Polanz; Peter Quehenberger; Christian Schörgenhofer; Johann Bartko; Robin Ristl; Bernd Jilma

Abstract Background and aims: The aim of this study was to assess the circadian variation and the between- and within-subject variation in 10 healthy subjects over a period of 8 weeks by ROTEM®. We further evaluated the influence of elevated body mass index and the effect of low molecular weight heparin and antithrombin on clot formation. Methods: Citrated blood samples were analysed in the NATEM® test system. The clotting time (CT), clot formation time (CFT), maximum clot firmness (MCF) and the maximum lysis (ML) were assessed. Results: Duplicate measurements showed that 23% of the CT and 31% of the CFT measurements had a coefficient of variation (CV) greater than 10%. The within-subject CV was 16% for the CT and 30% for the CFT. The MCF was fairly constant (6%), whereas ML showed more variation (18%). The between-subject CV was 6% for the CT and 20% for the CFT. Analytical variability was improved by summing up CT and CFT. Compared to morning values, CT, CFT and the sum of CT + CFT were shortened in the afternoon. High body mass index was associated with faster clotting. High concentrations of antithrombin had similar effects on clot formation as 0.2 IU/ml of enoxaparin. Conclusions: To overcome the influence of diurnal variation, we recommend obtaining blood samples at specified times in the morning. The within-subject variation should be taken into account, when serial measurements of drug effects are required.


Journal of Thrombosis and Haemostasis | 2016

Dexamethasone inhibits endotoxin-induced coagulopathy in human lungs.

Johann Bartko; Christian Schoergenhofer; Michael Schwameis; Nina Buchtele; Johann Wojta; Gernot Schabbauer; Leopold Stiebellehner; Bernd Jilma

Essentials Glucocorticoids are associated with an increased risk of thrombosis. Healthy volunteers received dexamethasone or placebo in an endotoxin lung instillation model. Dexamethasone suppressed thrombin generation in bronchoalveolar lavage. Glucocorticoids inhibit endotoxin induced pulmonary coagulopathy.

Collaboration


Dive into the Johann Bartko's collaboration.

Top Co-Authors

Avatar

Bernd Jilma

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Michael Schwameis

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Ulla Derhaschnig

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Quehenberger

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Christa Firbas

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Friedrich W. Kursten

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge