Network


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

Hotspot


Dive into the research topics where Peter Matzneller is active.

Publication


Featured researches published by Peter Matzneller.


Clinical Pharmacology & Therapeutics | 2012

Pgp-Mediated Interaction Between (R)-[11C]Verapamil and Tariquidar at the Human Blood–Brain Barrier: A Comparison With Rat Data

Martin Bauer; Markus Zeitlinger; Rudolf Karch; Peter Matzneller; Johann Stanek; Walter Jäger; M Böhmdorfer; Wolfgang Wadsak; Markus Mitterhauser; Jens P. Bankstahl; Wolfgang Löscher; Matthias J. Koepp; Claudia Kuntner; Markus Müller; Oliver Langer

Using positron emission tomography (PET) imaging we assessed, in vivo, the interaction between a microdose of (R)‐[11C]verapamil (a P‐glycoprotein (Pgp) substrate) and escalating doses of the Pgp inhibitor tariquidar (3, 4, 6, and 8 mg/kg) at the blood–brain barrier (BBB) in healthy human subjects. We compared the dose–response relationship of tariquidar in humans with data obtained in rats using a similar methodology. Tariquidar was equipotent in humans and rats in its effect of increasing (R)‐[11C]verapamil brain uptake (expressed as whole‐brain volume of distribution (VT)), with very similar half‐maximum‐effect concentrations. Both in humans and in rats, brain VT approached plateau levels at plasma tariquidar concentrations >1,000 ng/ml. However, Pgp inhibition in humans led to only a 2.7‐fold increase in brain VT relative to baseline scans (before administration of tariquidar) as compared with 11.0‐fold in rats. The results of this translational study add to the accumulating evidence that there are marked species‐dependent differences in Pgp expression and functionality at the BBB.


Antimicrobial Agents and Chemotherapy | 2013

Blood, tissue and intracellular concentrations of Azithromycin during and after end of therapy

Peter Matzneller; Shaip Krasniqi; Martina Kinzig; Fritz Sörgel; S. Hüttner; Edith Lackner; Markus Müller; Markus Zeitlinger

ABSTRACT Although azithromycin is extensively used in the treatment of respiratory tract infections as well as skin and skin-related infections, pharmacokinetics of azithromycin in extracellular space fluid of soft tissues, i.e., one of its therapeutic target sites, are not yet fully elucidated. In this study, azithromycin concentration-time profiles in extracellular space of muscle and subcutaneous adipose tissue, but also in plasma and white blood cells, were determined at days 1 and 3 of treatment as well as 2 and 7 days after the end of treatment. Of all compartments, azithromycin concentrations were highest in white blood cells, attesting for intracellular accumulation. However, azithromycin concentrations in both soft tissues were markedly lower than in plasma both during and after treatment. Calculation of the area under the concentration-time curve from 0 to 24 h (AUC0–24)/MIC90 ratios for selected pathogens suggests that azithromycin concentrations measured in the present study are subinhibitory at all time points in both soft tissues and at the large majority of observed time points in plasma. Hence, it might be speculated that azithromycins clinical efficacy relies not only on elevated intracellular concentrations but possibly also on its known pleotropic effects, including immunomodulation and influence on bacterial virulence factors. However, prolonged subinhibitory azithromycin concentrations at the target site, as observed in the present study, might favor the emergence of bacterial resistance and should therefore be considered with concern. In conclusion, this study has added important information to the pharmacokinetic profile of the widely used antibiotic drug azithromycin and evidentiates the need for further research on its potential for induction of bacterial resistance.


Journal of Antimicrobial Chemotherapy | 2011

The third-generation P-glycoprotein inhibitor tariquidar may overcome bacterial multidrug resistance by increasing intracellular drug concentration

I. Leitner; J. Nemeth; T. Feurstein; Aiman Abrahim; Peter Matzneller; H. Lagler; Thomas Erker; Oliver Langer; Markus Zeitlinger

OBJECTIVES The use of efflux pump inhibitors may be a powerful strategy to overcome transporter-mediated bacterial multidrug resistance. In the present study, we set out to investigate the potency of tariquidar, a third-generation P-glycoprotein inhibitor in clinical development, for overcoming bacterial resistance towards ciprofloxacin. METHODS Staphylococcus aureus 29213 (SA29213) and S. aureus 1199B (SA1199B), which overexpresses the multidrug transporter NorA, as well as Pseudomonas aeruginosa 27853 and Stenotrophomonas maltophilia BAA-85, which expresses SmeDEF, were exposed to ciprofloxacin in the presence and absence of tariquidar or, for comparative reasons, elacridar. Activity of both P-glycoprotein inhibitors was evaluated by determination of MICs and time-kill curves, and by quantification of uptake of ciprofloxacin into bacterial cells. RESULTS Activity of tariquidar and elacridar was comparable for S. aureus strains, and both dose-dependently increased susceptibility towards ciprofloxacin. Highest effects were observed for SA1199B, where the addition of tariquidar resulted in a 10-fold reduction of the ciprofloxacin MIC, while no effect was observed for P. aeruginosa. For S. maltophilia, elacridar but not tariquidar improved susceptibility. Uptake of [14C]ciprofloxacin and modification of susceptibility showed significant correlations (r=0.89, P<0.0001). Tariquidar had no intrinsic activity against any strain tested. CONCLUSIONS We conclude that tariquidar has potent inhibitory effect against certain bacterial efflux pumps in vitro. Their high activity at clinically achievable concentrations might yield this class of drugs promising for future applications in infectious diseases.


Journal of Antimicrobial Chemotherapy | 2016

Simultaneous assessment of the pharmacokinetics of a pleuromutilin, lefamulin, in plasma, soft tissues and pulmonary epithelial lining fluid.

Markus Zeitlinger; Richard Schwameis; Angela Burian; Bernhard Burian; Peter Matzneller; Markus Müller; W. W. Wicha; D. B. Strickmann; W. Prince

BACKGROUND Lefamulin is a pleuromutilin antibiotic under evaluation for the treatment of bacterial infections, including respiratory tract infections. Currently, there are no high-quality pharmacokinetic data on drug tissue concentrations of lefamulin available. METHODS A single dose of intravenous lefamulin (150 mg) was given to 12 healthy men. The registered EudraCT number for this study was 2010-021938-54. Lefamulin concentrations were simultaneously measured in plasma, skeletal muscle tissue, subcutaneous adipose tissue and epithelial lining fluid (ELF) over 24 h, and corresponding pharmacokinetic parameters were calculated. Microdialysis was used to measure unbound lefamulin concentrations in skeletal muscle tissue and subcutaneous adipose tissue, which were similar to unbound lefamulin concentrations in plasma. Bronchoalveolar lavage was performed 1, 2, 4 and 8 h post-dose to determine lefamulin concentrations in ELF. RESULTS Unbound lefamulin levels showed a 5.7-fold higher exposure in ELF compared with that in plasma, demonstrating good penetration to the target site. CONCLUSIONS Lefamulin may be an addition to the therapeutic armamentarium for the treatment of infections. Simultaneous measurements of unbound drug concentration can guide target attainment for future therapeutic trials.


Antimicrobial Agents and Chemotherapy | 2016

Single- and Repeated-Dose Pharmacokinetics of Ceftaroline in Plasma and Soft Tissues of Healthy Volunteers for Two Different Dosing Regimens of Ceftaroline Fosamil

Peter Matzneller; Edith Lackner; Heimo Lagler; Beatrix Wulkersdorfer; Zoe Österreicher; Markus Zeitlinger

ABSTRACT Ceftaroline fosamil (CPT-F) is currently approved for use for the treatment of complicated skin and soft tissue infections and community-acquired pneumonia at 600 mg twice daily (q12h), but other dosing regimens are under evaluation. To date, very limited data on the soft tissue pharmacokinetics (PK) of the active compound, ceftaroline (CPT), are available. CPT concentrations in the plasma, muscle, and subcutis of 12 male healthy volunteers were measured by microdialysis after single and repeated intravenous administration of 600 mg CPT-F q12h or three times daily (q8h) in two groups of 6 subjects each. Relevant PK and PK/pharmacodynamic (PD) parameters were calculated and compared between groups. In plasma, the area under the concentration-time curve (AUC) from 0 to 24 h for total CPT and the cumulative percentage of the dosing interval during which the free drug concentrations exceeded the MIC (fTMIC) for unbound CPT for the currently established threshold of 1 mg/liter were significantly higher in the group receiving CPT-F q8h. Exposure to free drug in soft tissues was higher in the group receiving CPT-F q8h, but high interindividual variability in relevant PK parameters was observed. The mean ratios of the AUC from time zero to the end of the dosing interval (AUC0-τ) for free CPT in soft tissues and the AUC0-τ for the calculated free fraction in plasma at steady state ranged from 0.66 to 0.75. Administration of CPT-F q8h led to higher levels of drug exposure in all investigated compartments. When MIC values above 1 mg/liter were assumed, the calculated fTMIC after dosing q12h was markedly lower than that after dosing q8h. The clinical implications of these differences are discussed in light of recently completed clinical phase III and PK/PD studies.


The Journal of Clinical Pharmacology | 2015

Feasibility of microdialysis for determination of protein binding and target site pharmacokinetics of colistin in vivo

Peter Matzneller; Patrice Gobin; Edith Lackner; Markus Zeitlinger

Tissue pharmacokinetics and plasma protein binding of colistin have not been described in humans in vivo. Colistin concentrations in plasma, muscle, and subcutis of healthy volunteers were measured by microdialysis after a single dose of 2.5 million IU of colistin methanesulfonate. In vitro microdialysis experiments and an in vivo pilot study were performed prior to the in vivo main study. Concentration‐time profiles of total colistin in plasma were comparable with previously described values. The unbound fraction of colistin in plasma (fu) ranged from 2.8% to 14.1%. Low plasma fu correlated with low unbound colistin concentrations in muscle and subcutis. In vitro, mean relative recovery of microdialysis probes was higher in the reverse dialysis setting compared to the forward dialysis mode (71 ± 9% vs. 45 ± 14%, respectively); mean overall recovery in the main study in vivo was 49 ± 5%. Present data suggest that colistin is extensively protein bound in plasma and poorly distributed into soft tissue. However, differences in relative recovery between forward and reverse dialysis in vitro indicate that results might have been influenced by adhesion of colistin to microdialysis equipment. Microdialysis should be considered as a semiquantitative method for the estimation of unbound colistin levels in soft tissue.


Antimicrobial Agents and Chemotherapy | 2012

Pharmacokinetics of Intraperitoneal and Intravenous Fosfomycin in Automated Peritoneal Dialysis Patients without Peritonitis

Selma Tobudic; Peter Matzneller; Brigitte Stoiser; Judith Maria Wenisch; Markus Zeitlinger; Andreas Vychytil; Walter Jaeger; Michaela Boehmdorfer; Gottfried Reznicek; Heinz Burgmann

ABSTRACT Blood and dialysate concentrations of fosfomycin were determined after intravenous and intraperitoneal application of 4 mg/liter in patients undergoing automated peritoneal dialysis. Maximum serum concentrations after intravenous (287.75 ± 86.34 mg/liter) and intraperitoneal (205.78 ± 66.78 mg/liter) administration were comparable. Ratios of intraperitoneal to systemic exposure were 1.12 (intraperitoneal administration) and 0.22 (intravenous administration), indicating good systemic exposure after intraperitoneal application but limited penetration of fosfomycin into the peritoneal fluid after the intravenous dose.


The Journal of Clinical Pharmacology | 2014

Target site pharmacokinetics of linezolid after single and multiple doses in diabetic patients with soft tissue infection

Roza Badr Eslam; Angela Burian; Greisa Vila; Robert Sauermann; Alexandra Hammer; Dorothea Frenzel; Iris K. Minichmayr; Charlotte Kloft; Peter Matzneller; Zoe Oesterreicher; Markus Zeitlinger

The underlying pathology of diabetic wounds, i.e. impairment of macro‐ and microcirculation, might also impact target site penetration of antibacterial drugs. To compare tissue concentrations of linezolid in infected and not infected tissue 10 patients suffering from type 2 diabetes with foot infection were included in the study. Tissue penetration of linezolid was assessed using in vivo microdialysis at the site of infection as well as in non‐inflamed subcutaneous adipose tissue. All patients were investigated after receiving a single dose of linezolid and five patients in addition at steady state. After a single dose of linezolid significantly higher area under the concentration vs. time curve over 8 hours (AUC0–8) and maximum concentrations (Cmax)‐values were observed in plasma (65.5 ± 21.2 mg*h/L and 16.4 ± 4.6 mg/L) as compared to inflamed (36.3 ± 22.9  mg*h/L and 6.6 ± 3.6 mg/L) and non‐inflamed tissue (33.0 ± 17.7 mg*h/L and 6.7 ± 3.6 mg/L). Multiple administrations of linezolid led to disappearance of significant differences in Cmax and AUC0–8 between plasma, inflamed, and non‐inflamed tissue. Approximately 2‐fold increase of Cmax and AUC0–8‐values in tissue was observed at steady state as compared to the first administration. Penetration of linezolid is not impaired in diabetic foot infection but equilibrium between plasma and tissue might be delayed.


Antimicrobial Agents and Chemotherapy | 2012

Blood, Tissue, and Intracellular Concentrations of Erythromycin and Its Metabolite Anhydroerythromycin during and after Therapy

Shaip Krasniqi; Peter Matzneller; Martina Kinzig; Fritz Sörgel; S. Hüttner; Edith Lackner; Markus Müller; Markus Zeitlinger

ABSTRACT For macrolides, clinical activity but also the development of bacterial resistance has been attributed to prolonged therapeutic and subtherapeutic concentrations. Although erythromycin is a long-established antimicrobial, concomitant determination of the pharmacokinetics of erythromycin and its metabolites in different compartments is limited. To better characterize the pharmacokinetics of erythromycin and its anhydrometabolite (anhydroerythromycin [AHE]) in different compartments during and after the end of treatment with 500 mg of erythromycin four times daily, concentration-time profiles were determined in plasma, interstitial space of muscle and subcutaneous adipose tissue, and white blood cells (WBCs) at days 1 and 3 of treatment and 2 and 7 days after end of therapy. In WBCs, concentrations of erythromycin exceeded those in plasma approximately 40-fold, while free concentrations in plasma and tissue were comparable. The observed delay of peak concentrations in tissue might be caused by fast initial cellular uptake. Two days after the end of treatment, subinhibitory concentrations were observed in plasma and interstitial space of both soft tissues, while 7 days after the end of treatment, erythromycin was not detectable in any compartment. This relatively short period of subinhibitory concentrations may be advantageous compared to other macrolides. The ratio of erythromycin over AHE on day 1 was highest in plasma (2.81 ± 3.45) and lowest in WBCs (0.27 ± 0.22). While the ratio remained constant between single dose and steady state, after the end of treatment the concentration of AHE declined more slowly than that of the parent compound, indicating the importance of the metabolite for the prolonged drug interaction of erythromycin.


Antimicrobial Agents and Chemotherapy | 2014

Development of a Population Pharmacokinetic Model Characterizing the Tissue Distribution of Azithromycin in Healthy Subjects

Songmao Zheng; Peter Matzneller; Markus Zeitlinger; Stephan Schmidt

ABSTRACT Recent clinical trials indicate that the use of azithromycin is associated with the emergence of macrolide resistance. The objective of our study was to simultaneously characterize free target site concentrations and correlate them with the MIC90s of clinically relevant pathogens. Azithromycin (500 mg once daily [QD]) was administered orally to 6 healthy male volunteers for 3 days. The free concentrations in the interstitial space fluid (ISF) of muscle and subcutaneous fat tissue as well as the total concentrations in plasma and polymorphonuclear leukocytes (PMLs) were determined on days 1, 3, 5, and 10. All concentrations were modeled simultaneously in NONMEM 7.2 using a tissue distribution model that accounts for nonlinear protein binding and ionization state at physiological pH. The model performance and parameter estimates were evaluated via goodness-of-fit plots and nonparametric bootstrap analysis. The model we developed described the concentrations at all sampling sites reasonably well and showed that the overall pharmacokinetics of azithromycin is driven by the release of the drug from acidic cell/tissue compartments. The model-predicted unionized azithromycin (AZM) concentrations in the cytosol of PMLs (6.0 ± 1.2 ng/ml) were comparable to the measured ISF concentrations in the muscle (8.7 ± 2.9 ng/ml) and subcutis (4.1 ± 2.4 ng/ml) on day 10, whereas the total PML concentrations were >1,000-fold higher (14,217 ± 2,810 ng/ml). The total plasma and free ISF concentrations were insufficient to exceed the MIC90s of the skin pathogens at all times. Our results indicate that the slow release of azithromycin from low pH tissue/cell compartments is responsible for the long terminal half-life of the drug and thus the extended period of time during which free concentrations reside at subinhibitory concentrations.

Collaboration


Dive into the Peter Matzneller's collaboration.

Top Co-Authors

Avatar

Markus Zeitlinger

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Markus Müller

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Edith Lackner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Angela Burian

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Oliver Langer

Austrian Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Zoe Österreicher

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Bauer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Shaip Krasniqi

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge