cellular accumulation (24h) Cstatic (LZD/RDZ ratio)

Slides:



Advertisements
Similar presentations
5/20/2015ISAP Is the cellular accumulation of antibiotics a predictive factor for intracellular efficacy ? Sandrine Lemaire Université catholique.
Advertisements

REVISED ABSTRACT Background: Omadacycline (OMC) is the first of a new class of tetracyclines, the aminomethylcyclines, and is being developed as a once-daily.
Hunting intracellular bacteria with antibiotics
Bacterial persistence
Isabelle K. Delattre, Françoise Van Bambeke and Paul M. Tulkens
Setting-up a model of intracellular infection by Pseudomonas aeruginosa for the pharmacodynamic evaluation of antibiotic activity. J. Buyck1, O. Jolois2,
studies with Caco-2 cells and THP-1 macrophages
Activities of Daptomycin (DAP), Vancomycin (VAN) and Linezolid (LDZ) alone or in combination with Fusidic acid (FUS) in an in vitro dynamic model of.
A-052 Antibiotic (AB) activity against Pseudomonas aeruginosa (PA) with Normal or Mucoïd Phenotypes in an Artificial Sputum Medium (ASM) in vitro Biofilm.
Activity of tobramycin in combination with clarithromycin
Introduction Results Aim of the study Methods References Conclusion
Intracellular Infection of THP-1 Human Macrophages by Staphylococcus aureus and Role of Haemolysins Mailling adresse: Aurélie Olivier.
Comparison of the intracellular and extracellular activities of approved and novel antistaphylococcal antibiotics using a pharmacodynamic model exploring.
Françoise Van Bambeke & Paul M. Tulkens
In vitro pharmacodynamic models for the study of antibiotic activity against bacterial biofilms Françoise Van Bambeke, PharmD, PhD Pharmacologie cellulaire.
Mix with THP-1 macrophages Overnight bacterial suspension
Frédéric Peyrusson, Françoise Van Bambeke, Paul M. Tulkens
Acknowledgments/Funding
Type III secretion system (TTSS) INTRODUCTION AND OBJECTIVES
Accumulation, Release and Subcellular Distribution of the Peptide Deformylase Inhibitor GSK in Murine Macrophages (J774) and Human Monocytes (THP-1)
In vitro susceptibility of S
Activity of 9 antibiotics against intracellular forms of S. pneumoniae
P1058 Intracellular activity of antibiotics against a stable small colony variant (SCV) isolated from a CF patient in a model Calu-3 human airway epithelial.
P 166 Introduction Description of the analyzed population
Revealing moxifloxacin activity against biofilms of S
Can efflux confer high levels resistance to meropenem (MEM) in Pseudomonas aeruginosa (Pa) clinical isolates? H. Chalhoub1, H. Rodriguez-Villalobos2,
Introduction & Purpose Results Conclusions
FUS, VAN and LZD injected twice (T0 and T12); DAP injected once (T0).
This poster will be made available for download after the meeting at :
Epithelial lining fluid penetration of temocillin administered by continuous infusion in critically ill patients with nosocomial pneumonia. Visée C.1a,
Introduction Results Aim Methods References Conclusion
Co-medications Improve Moxifloxacin (MXF) Activity in Models of Pneumococcal Naïve and Induced Biofilms (BF) N. M. Vandevelde1, M. Van Obbergh1, P.M. Tulkens1,
Background and objectives
Comparative in vitro activity of temocillin and other β-lactams
Acknowldgments and Funding
Activity and Pharmacodynamic (PD) Evaluation of Ceftazidime-Avibactam (CAZ-AVI) against Extracellular and Intracellular Forms of CAZ-susceptible and CAZ-resistant.
Inhibitors of type three secretion system [TTSS] protect against Pseudomonas aeruginosa cellular toxicity by inhibiting the transcription of TTSS Mailing.
Mapping French population [1]
Emilien Drouot, Paul M. Tulkens, Françoise Van Bambeke
Julien Buyck, Paul M. Tulkens and Françoise Van Bambeke
Julien Buyck, Paul M. Tulkens and Françoise Van Bambeke
Debaditya Das, Françoise Van Bambeke, Paul M. Tulkens
Extracellular activity Intracellular activity
Abstract Results Methods Background Conclusions References
UCL Université catholique de Louvain
Apoptosis induced by aminoglycosides (AGs) in cultured cells : comparison between gentamicin (GEN) and amikacin (AMK) using incubated and electroporated.
Incubation (with antibiotic)
52 - Antimicrobials Radezolid (RX-1741), a Novel Oxazolidinone, Accumulates Extensively within Human Macrophages and PMNs and Shows Activity towards.
Of a Pig-related ST-398 Methicillin-Resistant S. aureus (A-MRSA)
EV0626 Cellular pharmacokinetics of gepotidacin (GSK )
Abstract Results Aim of the study Methods Background Conclusions
Difference log CFU from time 0
DISCUSSION AND CONCLUSION
INTRODUCTION METHODS CONCLUSIONS REFERENCES
Introduction Results Materials and Methods Conclusions
1200 Brussels - Belgium Temocillin is not substrate for OprD2 porin from Pseudomonas aeruginosa H. Chalhoub1,
Electron microscopy: intracellular infection of THP-1 by S. aureus
Log extracellular concentration (mg/L)
Susceptibility of Pseudomonas aeruginosa (P. a
Activity of Moxifloxacin against Staphylococcus aureus in Models of Persistent Infections (Intracellular Survival, Biofilms) Mailing address: P.M. Tulkens.
M.P. Mingeot-Leclercq, P.M. Tulkens, F. Van Bambeke
Activities of antistaphylococcal antibiotics towards the extracellular and intraphagocytic forms of Staphylococcus aureus isolates from a patient with.
Françoise Van Bambeke, Dr Sc. Pharm, Agr. Ens. Sup.
Intracellular models of infection to evaluate antibiotic activity
Activities of antistaphylococcal antibiotics towards the extracellular and intraphagocytic forms of Staphylococcus aureus isolates from a patient with.
Setting-up of a 24 h model to evaluate the activity of antibiotics against intracellular forms of S. aureus infection C. Seral, M. Barcia-Macay, F. Van.
“Pulmonary infections”
The role of pharmacokinetics/pharmacodynamics in setting clinical MIC breakpoints: the EUCAST approach  J.W. Mouton, D.F.J. Brown, P. Apfalter, R. Cantón,
Are intracellular drug concentrations relevant for efficacy
Presentation transcript:

cellular accumulation (24h) Cstatic (LZD/RDZ ratio) Accumulation and Intracellular Activity of Radezolid, a New Oxazolidinone, in non-Phagocytic Cells S. Lemaire1, K. Kosowska-Shick2, P.C. Appelbaum2, J.-P. Pirnay,3 G. Verween,3 P. De Corte,3 D. De Vos,3 P.M. Tulkens1 and Françoise Van Bambeke1 Mailing address: F. Van Bambeke UCL 73.70 av. Mounier 73, 1200 Brussels Belgium francoise.vanbambeke@ulcouvain.be A1-1936 1 Université catholique de Louvain, Brussels, Belgium; 2 Hershey Medical Center, Hershey, PA, USA; Queen Astrid Military Hospital, Brussels, Belgium Abstract Results Background. S. aureus invades and persists within a variety of non-phagocytic cells. This may play a determining role in relapsing diseases like skin or respiratory tract infections, osteomyelitis, and endocarditis. Effective treatment may require drug accumulation in target cells. We have previously shown that radezolid (RDZ) accumulates 8-fold in human macrophages (Lemaire et al. ECCMID 2009, O29). We have now determined its intracell.. accumulation and activity in a series of relevant non-phagocytic cells. Methods. Cells are listed in the table. Cellular conc. was measured at 24h using 14C-RDZ, and accumulation calculated using a mean cell volume of 5 µl/mg cell prot. MICs and dose-effect relationships at 24h were determined as described (Barcia-Macay et al, AAC, 2006), using MRSA SA238 (clinical isolate) and a linezolid (LZD)R in vitro mutant thereof (SA238L). Results are expressed as the change in 24h inoculum compared to t=0. Results. The cellular accumulation of RDZ and its intracellular activity (against S. aureus strain SA238, [MICs: 0.5-1 and 2 mg/L for RDZ and LZD] was similar in all cell types. Against the LZDR mutant (MICs: 2 and 8 mg/L for RDZ and LZD), RDZ Cs and Emax remained constant while LZD activity was diminished (Cs range from 15.7 mg/L in Calu-3 to > 100 mg/L in osteoblasts; Emax, –0.6 log in keratinocytes to +0.5 log in osteoblasts). Conclusions. RDZ accumulates in non-phagocytic cells to the same extent as in macrophages and shows comparable intracellular activity against both LZDS and LZDR MRSA. LZD was systematically less effective against the resistant strain. Cellular accumulation of radezolid Intracellular activity of antibiotics Pharmacological descriptors as calculated by non-linear (sigmoid) regression of the dose-response curves illustrated in the Figure. Cell lines cellular accumulation (24h) Rat fibroblasts (primary culture) 10.9 ± 2.1 Human skin keratinocytes (primary cultures) 16.1 ± 2.0 Normal human osteoblasts (NHost) 9.7 ± 0.4 Human Umbilical Vein endothelial cells (HUVEC) 10.5 ± 0.1 Human lung epithelial cells (Calu-3) 8.3 ± 1.0 strain Cell line linezolid radezolid Cstatic (LZD/RDZ ratio) Csa (mg/L) Emaxb SA238 Fibroblasts ~ 6.6 -0.8 ± 0.1 ~ 1.5 -1.5 ± 0.3 4.4 (LNZ-S) Keratinocytes ~ 9.5 -1.2 ± 0.3 -1.2 ± 0.1 6.3 Osteoblasts ~ 6.8 -1.2 ± 0.2 ~ 1.3 -1.3 ± 0.1 5.2 HUVEC ~ 5.8 -0.5 ± 0.1 ~ 1.7 -0.7 ± 0.1 3.4 Calu-3 ~ 2.6 ~ 0.6 4.3 SA238L ~ 42 -0.2 ± 0.1 ~ 1.9 22 (LNZ-R) ~ 27 -0.6 ± 0.3 ~ 3.3 -1.3 ± 0.2 8.2 > 100 +0.5 ± 0.1 ~ 2.8 -1.0 ± 0.1 > 35 ~ 20 -0.4 ± 0.1 ~ 2.3 -0.6 ± 0.1 8.7 ~ 15 -0.2 ± 0.2 -1.4 ± 0.1 25 a Static concentration : concentration yielding no apparent change in the inoculum compared to time 0 b Maximal Efficacy: Change in log10 CFU compared to time 0 for an infinitely large concentration of antibiotic At 24 h, radezolid accumulates to a high and similar level within all non-phagocytic cells, reaching an accumulation factor similar to that observed within phagocytic cells (2). Concentration-killing effect of linezolid vs. radezolid against linezolid-susceptible SA238 and linezolid-resistant (SA238L) strains phagocytised by different cell types. Cells were incubated with antibiotics for 24 h in the presence of increasing concentrations of antibiotics. The graph shows the change in CFU per mg of cell protein. Background and aim Methods It is now well accepted that Staphylococcus aureus is a facultative intracellular pathogen and that its capacity to survive within non-phagocytic cells may play an important role in the persistent and recurrent character of several infections, such as complicated skin and soft tissue infections, osteomyelitis, endocarditis, or pulmonary infections associated with cystic fibrosis (1). Effective treatment of such infections may therefore require the use of antibiotics accumulating intracellularly and demonstrating activity therein. In this context, we have shown that radezolid (RDZ), a novel oxazolidinone antibiotic (see structure and description on poster A1-1937), markedly accumulates (approx. 10-fold) within phagocytic cells (PMN and macrophages) and shows improved potency towards both linezolid-susceptible and linezolid-resistant S. aureus phagocytized by human THP-1 macrophages (2; A1-1937). The aim of the present study was to measure the intracellular accumulation and activity of radezolid in a series of non-phagocytic cells originating from the tissues subject to persistent infections. Bacteria and susceptibility testings. S. aureus SA238 and SA238L, a linezolid-resistant mutant thereof selected in vitro, were used in the present study (3). MICs were determined in Mueller Hinton Broth as previously described (4). Cell lines. Experiments were conducted with promiary cultures of rat fibroblasts or of human skin keratinocytes, and with continous cell lines of human lung epithelial cells (ATCC, Manassas, VA), normal human osteoblasts (Lonza), and human umbilical Vein endothelial cells (HUVEC, Lonza). Determination of the cellular accumulation. The cellular content in radezolid was determined by scintillation, cells having been incubated with 14C-RDZ. Cell infection and assessment of the intracellular activities of antibiotics. Infection of cells was performed exactly as described before (3-5) with typical starting inoculum of ~ 1 to 2x106 CFU/mg prot. Antibiotic activitiy was examined after 24 h in the presence of increasing concentration of antibiotics (0.01- to 100 mg/L), and results were expressed as the change in the inoculum at 24 h compared to time 0 (4-5). Data were analyzed by non-linear regression using Hill's equation to calculate pharmacological descriptors (Emax, maximal reduction of the intracellular inoculum [in log10 units] for an infinitely large antibiotic concentration; EC50, antibiotic concentration [in mg/L] yielding a response half-way between E0 and Emax, as obtained by graphical intrapolation using the corresponding Hill equation), together with an estimation of the static concentration. Radezolid shows similar efficacy (Emax) and potency (Cstatic) against both the SA238 susceptible and the SA238L resistant strains. Radezolid is 3.4- to 6.3-fold more potent than linezolid against the SA238 susceptible strain and 8.7 to > 100 fold against the SA238L resistant strain. Although MICs are the same (2 mg/L) , radezolid shows 2-3-fold higher potency against the resistant strain SA238L than linezolid against the susceptible strain SA238. Radezolid shows similar efficacy in all cell types ; linezolid efficacy is reduced against the SA238L resistant strain in all cell lines except HUVEC. Conclusions References Radezolid accumulates to a similar extent in all cell types . Radezolid shows globally a similar relative efficacy (Emax) than linezolid but a higher relative potency (lower Cs) than linezolid against intracellular S. aureus in all cell types. Radezolid shows similar intracellular activities (Emax and Cs) against linezolid-susceptible and linezolid resistant strains, suggesting that it could be a useful substitute to linezolid to treat intracellular infections caused by these resistant bacteria. Garzoni & Kelley. Trends Microbiol. (2009) 17:59-65. Lemaire et al. 26th ICC, Toronto, ON (2009) Kosowska-Shick et al. Antimicrob. Agents Chemother. (2006) 50:765-769. Barcia-Macay et al. Antimicrob. Agents Chemother. (2006) 50: 841-851. Lemaire et al, Antimicrob. Agents Chemother. (2008) 52:2797-2805. This poster will be made available for download after the meeting: http://www.facm.ucl.ac.be/posters.htm We thank M.C. Cambier and C. Misson for technical assistance. S.L. and F.V.B. are Post-doctoral and Senior Research Associate of the Belgian Fonds de la Recherche Scientifique (FSR-FNRS). This work was supported by the Belgian Fonds de la Recherche Scientifique Médicale (FRSM) and a grant-in-aid from Rib-X Pharmaceuticals.