99 results on '"Arnold Louie"'
Search Results
2. 1695. Combination Therapy for the Investigational Polymyxin SPR206 and Meropenem (MEM) Increases the Rapidity and Extent of Killing of Pseudomonas aeruginosa (PA) and Prevents the Bacterium from Emerging Resistant to Both Antibiotics
- Author
-
Arnold Louie, Justin Bader, Nicole Cotroneo, Troy Lister, Weiguo Liu, David Brown, Jenny Myrick, and George Drusano
- Subjects
Infectious Diseases ,Oncology - Abstract
Background The CDC lists multidrug-resistant PA as a Serious Threat pathogen causing 32,200 hospital infections and 2,700 deaths in the US in 2017. PA resistance is frequent with antibiotic monotherapies. SPR206 is a next generation polymyxin that, in a first in human study, was generally safe and well tolerated at the potential therapeutic dose of 100 mg IV Q8h. The aim of this study was to quantify the dose-range PA killing activity of SPR206 monotherapy in a hollow fiber infection model (HFIM) and to assess if the combination of SPR206 and MEM more effectively reduces bacterial burden and prevents development of resistance than either drug alone. Methods Agar dilution MICs and mutation frequencies (MFs) for SPR206, PMB, & MEM were performed for PA ATCC 27853. A 10-day HFIM dose range study simulating the free PK profiles for SPR206 (50 – 600 mg IV Q8h) quantified PA killing and possible regrowth. A HFIM study compared single vs combination SPR206 100 mg IV Q8h over 1h and MEM 2g IV Q8h over 1h regimens for enhanced PA killing and resistance suppression. Results The SPR206, PMB, & MEM MICs were 0.5, 0.5, and 0.25 mg/L, respectively. The MFs for SPR206 and PMB to 3x, 5x and 8x MIC were -6.2, -6.9, & -7.7 log CFU and -5.3, -6.4, & -7.7 log CFU, respectively. The MF for 3x MIC of MEM was -6.8 log CFU. In a dose-range HFIM, simulated regimens for 50 – 600 mg IV Q8h showed a dose-response effect, with 2.9 – 7.3 log CFU/mL reductions in PA seen at 5h. All regimens had regrowth by isolates with SPR206 MICs of 1 – 8 mg/L. In a HFIM study simulating the SPR206 and MEM clinical regimens, alone and in combination, SPR206 alone killed 5 log CFU/mL of PA at 5h, followed by regrowth. MEM alone killed 3.5 log CFU/mL of PA at 5h, with maximum kill seen on Day 4, followed by regrowth. SPR206 plus MEM killed 0.8 log CFU/mL more PA at 5h vs SPR206 alone and had undetectable PA counts by day 4. Combination therapy prevented regrowth (see figure). Conclusion Conclusions: SPR206 (100 mg IV Q8h) killed 5 log CFU/mL of PA at 5h, but regrowth ensued. SPR206 plus MEM produced 0.8 log CFU/mL more killing of PA at 5h. The killing activity of the 2-drug regimen combined with its resistance prevention effect resulted in undetectable PA counts by Day 4 of treatment. Disclosures Arnold Louie, MD, Curza, Inc.: Grant/Research Support|Prokaryotics: Grant/Research Support|Spero Therapeutics: Grant/Research Support Nicole Cotroneo, B.S., Spero Therapeutics: Employee|Spero Therapeutics: Stocks/Bonds Weiguo Liu, M.D., Spero Therapeutics: Grant/Research Support David Brown, M.B.A., Curza, Inc.: Grant/Research Support|Prokaryotics: Grant/Research Support|Spero Therapeutics: Grant/Research Support Jenny Myrick, B.S., Spero Therapeutics: Grant/Research Support George Drusano, M.D., Curza: Advisor/Consultant|Curza: Grant/Research Support|Prokaryotics: Grant/Research Support|Spero Therapeutics: Grant/Research Support.
- Published
- 2022
3. Pharmacodynamic evaluation of lefamulin in the treatment of gonorrhea using a hollow fiber infection model simulating Neisseria gonorrhoeae infections
- Author
-
Susanne Jacobsson, Daniel Golparian, Joakim Oxelbark, Wolfgang W. Wicha, Renata Maria Augusto da Costa, Francois Franceschi, David Brown, Arnold Louie, Steven P. Gelone, George Drusano, and Magnus Unemo
- Subjects
Pharmacology ,Pharmacology (medical) - Abstract
The emergence and spread of antimicrobial resistance in Neisseria gonorrhoeae is seriously threatening the treatment and control of gonorrhea globally. Novel treatment options are essential, coupled with appropriate methods to pharmacodynamically examine the efficacy and resistance emergence of these novel drugs. Herein, we used our dynamic in vitro hollow fiber infection model (HFIM) to evaluate protein-unbound lefamulin, a semisynthetic pleuromutilin, against N. gonorrhoeae. Dose–range and dose–fractionation experiments with N. gonorrhoeae reference strains: WHO F (susceptible to all relevant antimicrobials), WHO X (extensively drug-resistant, including ceftriaxone resistance), and WHO V (high-level azithromycin resistant, and highest gonococcal MIC of lefamulin (2 mg/l) reported), were performed to examine lefamulin gonococcal killing and resistance development during treatment. The dose–range experiments, simulating a single oral dose of lefamulin based on human plasma concentrations, indicated that ≥1.2 g, ≥2.8 g, and ≥9.6 g of lefamulin were required to eradicate WHO F, X, and V, respectively. Dose–fractionation experiments, based on human lefamulin plasma concentrations, showed that WHO X was eradicated with ≥2.8 g per day when administered as q12 h (1.4 g twice a day) and with ≥3.6 g per day when administered as q8 h (1.2 g thrice a day), both for 7 days. However, when simulating the treatment with 5–10 times higher concentrations of free lefamulin in relevant gonorrhea tissues (based on urogenital tissues in a rat model), 600 mg every 12 h for 5 days (approved oral treatment for community-acquired bacterial pneumonia) eradicated all strains, and no lefamulin resistance emerged in the successful treatment arms. In many arms failing single or multiple dose treatments for WHO X, lefamulin-resistant mutants (MIC = 2 mg/l), containing an A132V amino acid substitution in ribosomal protein L3, were selected. Nevertheless, these lefamulin-resistant mutants demonstrated an impaired biofitness. In conclusion, a clinical study is warranted to elucidate the clinical potential of lefamulin as a treatment option for uncomplicated gonorrhea (as well as several other bacterial STIs).
- Published
- 2022
4. Combination Therapy to Kill Mycobacterium tuberculosis in Its Nonreplicating Persister Phenotype
- Author
-
Walter Yamada, Sarah Kim, Mohammed Almoslem, Soyoung Kim, Jenny Myrick, Jocelyn Nole, Brandon Duncanson, Arnold Louie, Charles A. Peloquin, Stephan Schmidt, George L. Drusano, and Michael Neely
- Subjects
Pharmacology ,Drug Combinations ,Phenotype ,Infectious Diseases ,Moxifloxacin ,Antitubercular Agents ,Animals ,Humans ,Pharmacology (medical) ,Mycobacterium tuberculosis - Abstract
Mycobacterium tuberculosis (Mtb) exists in various metabolic states, including a nonreplicating persister (NRP) phenotype which may affect response to therapy. We have adopted a model-informed strategy to accelerate discovery of effective Mtb treatment regimens and previously found pretomanid (PMD), moxifloxacin (MXF), and bedaquiline (BDQ) to readily kill logarithmic- and acid-phase Mtb. Here, we studied multiple concentrations of each drug in flask-based, time-kill studies against NRP Mtb in single-, two- and three-drug combinations, including the active M2 metabolite of BDQ. We used nonparametric population algorithms in the Pmetrics package for R to model the data and to simulate the 95% confidence interval of bacterial population decline due to the two-drug combination regimen of PMD + MXF and compared this to observed declines with three-drug regimens. PMD + MXF at concentrations equivalent to average or peak human concentrations effectively eradicated Mtb. Unlike other states for Mtb, we observed no sustained emergence of less susceptible isolates for any regimen. The addition of BDQ as a third drug significantly (P < 0.05) shortened time to total bacterial suppression by 3 days compared to the two-drug regimen, similar to our findings for Mtb in logarithmic or acid growth phases.
- Published
- 2022
5. Pharmacodynamic evaluation of lefamulin in the treatment of gonorrhea using a hollow fiber infection model simulating
- Author
-
Susanne, Jacobsson, Daniel, Golparian, Joakim, Oxelbark, Wolfgang W, Wicha, Renata Maria Augusto, da Costa, Francois, Franceschi, David, Brown, Arnold, Louie, Steven P, Gelone, George, Drusano, and Magnus, Unemo
- Abstract
The emergence and spread of antimicrobial resistance in
- Published
- 2022
6. Combating Multidrug‐Resistant Bacteria by Integrating a Novel Target Site Penetration and Receptor Binding Assay Platform Into Translational Modeling
- Author
-
Deanna Deveson Lucas, Cornelia B. Landersdorfer, Jürgen B. Bulitta, Tae Hwan Kim, Dhruvitkumar S. Sutaria, Amy Wright, John D. Boyce, Arnold Louie, Alicja J. Copik, Brian T. Tsuji, Herbert P. Schweizer, Michael H. Norris, Miranda J. Wallace, Jeremiah Oyer, Carolin Werkman, Suresh Dharuman, Eric D. LoVullo, Beom Soo Shin, Yinzhi Lang, Xun Tao, Rossie H. Jimenez-Nieves, Brett A. Fleischer, Yuanyuan Jiao, Robert A. Bonomo, Venkataraman Balasubramanian, Kari B. Basso, Eunjeong Shin, Marianne Mégroz, George L. Drusano, Victoria C. Loudon-Hossler, Keisha C. Cadet, Nagakumar Bharatham, Jieqiang Zhou, Stephanie M. Reeve, Bartolomé Moyá, Nirav Shah, Alaa R. M. Sayed, and Richard E. Lee
- Subjects
medicine.drug_class ,Antibiotics ,Computational biology ,beta-Lactams ,Proteomics ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,Drug Resistance, Multiple, Bacterial ,Drug Discovery ,Gram-Negative Bacteria ,medicine ,Animals ,Humans ,Penicillin-Binding Proteins ,Pharmacology (medical) ,Multiplex ,Pharmacology ,Bacteriological Techniques ,biology ,Chemistry ,Cell Membrane ,Periplasmic space ,Models, Theoretical ,biology.organism_classification ,Disease Models, Animal ,Drug development ,030220 oncology & carcinogenesis ,Bacterial outer membrane ,Bacteria ,Systems pharmacology - Abstract
Multidrug-resistant bacteria are causing a serious global health crisis. A dramatic decline in antibiotic discovery and development investment by pharmaceutical industry over the last decades has slowed the adoption of new technologies. It is imperative that we create new mechanistic insights based on latest technologies, and use translational strategies to optimize patient therapy. Although drug development has relied on minimal inhibitory concentration testing and established in vitro and mouse infection models, the limited understanding of outer membrane permeability in Gram-negative bacteria presents major challenges. Our team has developed a platform using the latest technologies to characterize target site penetration and receptor binding in intact bacteria that inform translational modeling and guide new discovery. Enhanced assays can quantify the outer membrane permeability of β-lactam antibiotics and β-lactamase inhibitors using multiplex liquid chromatography tandem mass spectrometry. While β-lactam antibiotics are known to bind to multiple different penicillin-binding proteins (PBPs), their binding profiles are almost always studied in lysed bacteria. Novel assays for PBP binding in the periplasm of intact bacteria were developed and proteins identified via proteomics. To characterize bacterial morphology changes in response to PBP binding, high-throughput flow cytometry and time-lapse confocal microscopy with fluorescent probes provide unprecedented mechanistic insights. Moreover, novel assays to quantify cytosolic receptor binding and intracellular drug concentrations inform target site occupancy. These mechanistic data are integrated by quantitative and systems pharmacology modeling to maximize bacterial killing and minimize resistance in in vitro and mouse infection models. This translational approach holds promise to identify antibiotic combination dosing strategies for patients with serious infections.
- Published
- 2021
7. Pharmacodynamic Evaluation of Zoliflodacin Treatment of Neisseria gonorrhoeae Strains With Amino Acid Substitutions in the Zoliflodacin Target GyrB Using a Dynamic Hollow Fiber Infection Model
- Author
-
Susanne Jacobsson, Daniel Golparian, Joakim Oxelbark, Francois Franceschi, David Brown, Arnold Louie, George Drusano, and Magnus Unemo
- Subjects
Pharmacology ,Pharmacology (medical) - Abstract
Novel antimicrobials for effective treatment of uncomplicated gonorrhea are essential, and the first-in-class, oral spiropyrimidinetrione DNA gyrase B inhibitor zoliflodacin appears promising. Using our newly developed Hollow Fiber Infection Model (HFIM), the pharmacodynamics of zoliflodacin was examined. A clinical zoliflodacin-susceptible N. gonorrhoeae strain, SE600/18 (harbouring a GyrB S467N amino acid substitution; MIC = 0.25 mg/L), and SE600/18-D429N (zoliflodacin-resistant mutant with a second GyrB substitution, D429N, selected in the HFIM experiments; zoliflodacin MIC = 2 mg/L), were examined. Dose-range experiments, simulating zoliflodacin single oral dose regimens of 0.5, 1, 2, 3, and 4 g, were performed for SE600/18. For SE600/18-D429N, dose-range experiments, simulating zoliflodacin single oral 2, 3, 4, and 6 g doses, and zoliflodacin oral dose-fractionation experiments with 4, 6, and 8 g administered as q12 h were performed. Both strains grew well in the untreated HFIM growth control arms and mostly maintained growth at 1010–1011 CFU/ml for 7 days. Zoliflodacin 3 and 4 g single dose oral regimens successfully eradicated SE600/18 and no growth was recovered during the 7-days experiments. However, the single oral 0.5, 1, and 2 g doses failed to eradicate SE600/18, and zoliflodacin-resistant populations with a GyrB D429N substitution were selected with all these doses. The zoliflodacin-resistant SE600/18-D429N mutant was not eradicated with any examined treatment regimen. However, this in vitro-selected zoliflodacin-resistant mutant was substantially less fit compared to the zoliflodacin-susceptible SE600/18 parent strain. In conclusion, the rare clinical gonococcal strains with GyrB S467N substitution are predisposed to develop zoliflodacin resistance and may require treatment with zoliflodacin ≥3 g. Future development may need to consider the inclusion of diagnostics directed at identifying strains resistant or predisposed to resistance development at a population level and to strengthen surveillance (phenotypically and genetically), and possibly also at the patient level to guide treatment.
- Published
- 2022
8. Pharmacodynamic Evaluation of Zoliflodacin Treatment of
- Author
-
Susanne, Jacobsson, Daniel, Golparian, Joakim, Oxelbark, Francois, Franceschi, David, Brown, Arnold, Louie, George, Drusano, and Magnus, Unemo
- Abstract
Novel antimicrobials for effective treatment of uncomplicated gonorrhea are essential, and the first-in-class, oral spiropyrimidinetrione DNA gyrase B inhibitor zoliflodacin appears promising. Using our newly developed Hollow Fiber Infection Model (HFIM), the pharmacodynamics of zoliflodacin was examined. A clinical zoliflodacin-susceptible
- Published
- 2022
9. Building Optimal Three-Drug Combination Chemotherapy Regimens To Eradicate Mycobacterium tuberculosis in Its Slow-Growth Acid Phase
- Author
-
Michael Neely, Charles A. Peloquin, Sarah Parker, Arnold Louie, Jocelyn Nole, Sarah Kim, Stephanie Rogers, Stephan Schmidt, Nino Mtchedlidze, George L. Drusano, Brandon Duncanson, Walter M. Yamada, and Meredith Bacci
- Subjects
Tuberculosis ,Combination therapy ,Moxifloxacin ,Antitubercular Agents ,Pharmacology ,Mycobacterium tuberculosis ,chemistry.chemical_compound ,Animals ,Humans ,Medicine ,Pharmacology (medical) ,biology ,business.industry ,Combination chemotherapy ,medicine.disease ,biology.organism_classification ,Drug Combinations ,Regimen ,Infectious Diseases ,chemistry ,Pretomanid ,Drug Therapy, Combination ,Bedaquiline ,business ,medicine.drug - Abstract
Mycobacterium tuberculosis metabolic state affects the response to therapy. Quantifying the effect of antimicrobials in the acid and nonreplicating metabolic phases of M. tuberculosis growth will help to optimize therapy for tuberculosis. As a brute-force approach to all possible drug combinations against M. tuberculosis in all different metabolic states is impossible, we have adopted a model-informed strategy to accelerate the discovery. Using multiple concentrations of each drug in time-kill studies, we examined single drugs and two- and three-drug combinations of pretomanid, moxifloxacin, and bedaquiline plus its active metabolite against M. tuberculosis in its acid-phase metabolic state. We used a nonparametric modeling approach to generate full distributions of interaction terms between pretomanid and moxifloxacin for susceptible and less susceptible populations. From the model, we could predict the 95% confidence interval of the simulated total bacterial population decline due to the 2-drug combination regimen of pretomanid and moxifloxacin and compare this to observed declines with 3-drug regimens. We found that the combination of pretomanid and moxifloxacin at concentrations equivalent to average or peak human concentrations effectively eradicated M. tuberculosis in its acid growth phase and prevented emergence of less susceptible isolates. The addition of bedaquiline as a third drug shortened time to total and less susceptible bacterial suppression by 8 days compared to the 2-drug regimen, which was significantly faster than the 2-drug kill.
- Published
- 2021
10. Evaluating the effect of clofazimine against Mycobacterium tuberculosis given alone or in combination with pretomanid, bedaquiline or linezolid
- Author
-
Sarah Kim, Arnold Louie, George L. Drusano, Mohammed Almoslem, Soyoung Kim, Jenny Myrick, Jocelyn Nole, Brandon Duncanson, Charles A. Peloquin, Charles A. Scanga, Walter Yamada, Michael Neely, and Stephan Schmidt
- Subjects
Microbiology (medical) ,Infectious Diseases ,Nitroimidazoles ,Tuberculosis, Multidrug-Resistant ,Antitubercular Agents ,Linezolid ,Humans ,Pharmacology (medical) ,General Medicine ,Microbial Sensitivity Tests ,Mycobacterium tuberculosis ,Diarylquinolines ,Clofazimine - Abstract
In recent years, clofazimine (CFZ) has been regaining prominence for the treatment of tuberculosis. However, it shows limited efficacy as a single drug and optimal combination partners have not been identified. Therefore, the objective of our analysis was to evaluate the efficacy of CFZ-containing two-drug regimens with pretomanid (PMD), bedaquiline (BDQ) or linezolid (LZD) by: (i) determining their pharmacodynamic (PD) mode of interaction against Mycobacterium tuberculosis (Mtb) strain H37Rv in log- phase and acid-phase metabolic states, and against Mtb strain 18b in a non-replicating persister (NRP) metabolic state; (ii) predicting bacterial cell kill of the drugs alone and in combination; and (iii) evaluating the relationship between the interaction mode and the extent of bacterial cell kill. The results of our Greco universal response surface analysis showed that CFZ was at least additive with a clear trend towards synergy when combined with PMD, BDQ and LZD against Mtb in all explored metabolic states under in vitro checkerboard assay conditions. The results further showed that all two-drug combination regimens exerted greater bacterial kill than any of the drugs alone. CFZ alone showed the least antimicrobial efficacy amongst the evaluated drugs, and there was a lack of correlation between the mode of interaction and the extent of bacterial kill. However, we may underestimate the effect of CFZ in this screening approach owing to limited in vitro study duration and neglect of target site accumulation. Clofazimine; Pretomanid; Bedaquiline; Linezolid; Combination chemotherapy; Mycobacterium tuberculosis.
- Published
- 2021
11. Emergence of Resistance to Ceftazidime-Avibactam in a Pseudomonas aeruginosa Isolate Producing Derepressed bla PDC in a Hollow-Fiber Infection Model
- Author
-
Laura J. Rojas, Mark Raymond Adams, Jürgen B. Bulitta, Michael Vicchiarelli, Maria F. Mojica, Liang Chen, Barry N. Kreiswirth, Robert A. Bonomo, Steven M Marshall, Meredith Bacci, Nino Mtchedlidze, George L. Drusano, and Arnold Louie
- Subjects
Pharmacology ,0303 health sciences ,Deletion mutant ,030306 microbiology ,Pseudomonas aeruginosa ,Chemistry ,Continuous infusion ,Mutant ,Ceftazidime ,Liter ,medicine.disease_cause ,Ceftazidime/avibactam ,Microbiology ,03 medical and health sciences ,Infectious Diseases ,medicine ,Pharmacology (medical) ,030304 developmental biology ,medicine.drug - Abstract
Ceftazidime (CAZ)-avibactam (AVI) is a β-lactam/β-lactamase inhibitor combination with activity against type A and type C β-lactamases. Resistance emergence has been seen, with multiple mechanisms accounting for the resistance. We performed four experiments in the dynamic hollow-fiber infection model, delineating the linkage between drug exposure and both the rate of bacterial kill and resistance emergence by all mechanisms. The Pseudomonas aeruginosa isolate had MICs of 1.0 mg/liter (CAZ) and 4 mg/liter (AVI). We demonstrated that the time at ≥4.0 mg/liter AVI was linked to the rate of bacterial kill. Linkage to resistance emergence/suppression was more complex. In one experiment in which CAZ and AVI administration was intermittent and continuous, respectively, and in which AVI was given in unitary steps from 1 to 8 mg/liter, AVI at up to 3 mg/liter allowed resistance emergence, whereas higher values did not. The threshold value was 3.72 mg/liter as a continuous infusion to counterselect resistance (AVI area under the concentration-time curve [AUC] of 89.3 mg · h/liter). The mechanism involved a 7-amino-acid deletion in the Ω-loop region of the Pseudomonas-derived cephalosporinase (PDC) β-lactamase. Further experiments in which CAZ and AVI were both administered intermittently with regimens above and below the AUC of 89.3 mg · h/liter resulted in resistance in the lower-exposure groups. Deletion mutants were not identified. Finally, in an experiment in which paired exposures as both continuous and intermittent infusions were performed, the lower value of 25 mg · h/liter by both profiles allowed selection of deletion mutants. Of the five instances in which these mutants were recovered, four had a continuous-infusion profile. Both continuous-infusion administration and low AVI AUC exposures have a role in selection of this mutation.
- Published
- 2021
12. Emergence of Resistance to Ceftazidime-Avibactam in a Pseudomonas aeruginosa Isolate Producing Derepressed
- Author
-
G L, Drusano, Robert A, Bonomo, Steven M, Marshall, Laura J, Rojas, Mark D, Adams, Maria F, Mojica, Barry N, Kreiswirth, Liang, Chen, Nino, Mtchedlidze, Meredith, Bacci, Michael, Vicchiarelli, Jürgen B, Bulitta, and Arnold, Louie
- Subjects
Drug Combinations ,Pseudomonas ,Pseudomonas aeruginosa ,Experimental Therapeutics ,Microbial Sensitivity Tests ,Azabicyclo Compounds ,Ceftazidime ,Anti-Bacterial Agents ,Cephalosporinase - Abstract
Ceftazidime (CAZ)-avibactam (AVI) is a β-lactam/β-lactamase inhibitor combination with activity against type A and type C β-lactamases. Resistance emergence has been seen, with multiple mechanisms accounting for the resistance. We performed four experiments in the dynamic hollow-fiber infection model, delineating the linkage between drug exposure and both the rate of bacterial kill and resistance emergence by all mechanisms. The Pseudomonas aeruginosa isolate had MICs of 1.0 mg/liter (CAZ) and 4 mg/liter (AVI). We demonstrated that the time at ≥4.0 mg/liter AVI was linked to the rate of bacterial kill. Linkage to resistance emergence/suppression was more complex. In one experiment in which CAZ and AVI administration was intermittent and continuous, respectively, and in which AVI was given in unitary steps from 1 to 8 mg/liter, AVI at up to 3 mg/liter allowed resistance emergence, whereas higher values did not. The threshold value was 3.72 mg/liter as a continuous infusion to counterselect resistance (AVI area under the concentration-time curve [AUC] of 89.3 mg · h/liter). The mechanism involved a 7-amino-acid deletion in the Ω-loop region of the Pseudomonas-derived cephalosporinase (PDC) β-lactamase. Further experiments in which CAZ and AVI were both administered intermittently with regimens above and below the AUC of 89.3 mg · h/liter resulted in resistance in the lower-exposure groups. Deletion mutants were not identified. Finally, in an experiment in which paired exposures as both continuous and intermittent infusions were performed, the lower value of 25 mg · h/liter by both profiles allowed selection of deletion mutants. Of the five instances in which these mutants were recovered, four had a continuous-infusion profile. Both continuous-infusion administration and low AVI AUC exposures have a role in selection of this mutation.
- Published
- 2021
13. First Penicillin-Binding Protein Occupancy Patterns for 15 β-Lactams and β-Lactamase Inhibitors in Mycobacterium abscessus
- Author
-
Xun Tao, Alaa R. M. Sayed, Jieqiang Zhou, Arnold Louie, Eunjeong Shin, Weiguo Liu, Yinzhi Lang, Carolin Werkman, Dhruvitkumar S. Sutaria, Nirav Shah, George L. Drusano, Bartolomé Moyá, Yuanyuan Jiao, Manasi Kamat, Kari B. Basso, and Jürgen B. Bulitta
- Subjects
Cefotaxime ,Penicillin binding proteins ,Avibactam ,Microbial Sensitivity Tests ,Penicillins ,Aztreonam ,Mycobacterium abscessus ,beta-Lactams ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,polycyclic compounds ,medicine ,Penicillin-Binding Proteins ,Pharmacology (medical) ,Cefoxitin ,Mecillinam ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,030306 microbiology ,Chemistry ,Sulbactam ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,Anti-Bacterial Agents ,Infectious Diseases ,bacteria ,beta-Lactamase Inhibitors ,medicine.drug - Abstract
Mycobacterium abscessus causes serious infections that often require over 18 months of antibiotic combination therapy. There is no standard regimen for the treatment of M. abscessus infections, and the multitude of combinations that have been used clinically have had low success rates and high rates of toxicities. With β-lactam antibiotics being safe, double β-lactam and β-lactam/β-lactamase inhibitor combinations are of interest for improving the treatment of M. abscessus infections and minimizing toxicity. However, a mechanistic approach for building these combinations is lacking since little is known about which penicillin-binding protein (PBP) target receptors are inactivated by different β-lactams in M. abscessus. We determined the preferred PBP targets of 13 β-lactams and 2 β-lactamase inhibitors in two M. abscessus strains and identified PBP sequences by proteomics. The Bocillin FL binding assay was used to determine the β-lactam concentrations that half-maximally inhibited Bocillin binding (50% inhibitory concentrations [IC(50)s]). Principal component analysis identified four clusters of PBP occupancy patterns. Carbapenems inactivated all PBPs at low concentrations (0.016 to 0.5 mg/liter) (cluster 1). Cephalosporins (cluster 2) inactivated PonA2, PonA1, and PbpA at low (0.031 to 1 mg/liter) (ceftriaxone and cefotaxime) or intermediate (0.35 to 16 mg/liter) (ceftazidime and cefoxitin) concentrations. Sulbactam, aztreonam, carumonam, mecillinam, and avibactam (cluster 3) inactivated the same PBPs as cephalosporins but required higher concentrations. Other penicillins (cluster 4) specifically targeted PbpA at 2 to 16 mg/liter. Carbapenems, ceftriaxone, and cefotaxime were the most promising β-lactams since they inactivated most or all PBPs at clinically relevant concentrations. These first PBP occupancy patterns in M. abscessus provide a mechanistic foundation for selecting and optimizing safe and effective combination therapies with β-lactams.
- Published
- 2020
14. Developing New Drugs for Mycobacterium tuberculosis Therapy: What Information Do We Get from Preclinical Animal Models?
- Author
-
Charles A. Scanga, Charles A. Peloquin, Arnold Louie, Michael Vicchiarelli, Brandon Duncanson, Michael Neely, Walter M. Yamada, Stephan Schmidt, George L. Drusano, and Sarah Kim
- Subjects
Tuberculosis ,Antitubercular Agents ,Pharmacology ,Mycobacterium tuberculosis ,Mice ,03 medical and health sciences ,Cmin ,chemistry.chemical_compound ,Animal model ,Pharmacokinetics ,Animals ,Medicine ,Experimental Therapeutics ,Pharmacology (medical) ,Prospective Studies ,030304 developmental biology ,0303 health sciences ,biology ,030306 microbiology ,business.industry ,Epithelial lining fluid ,biology.organism_classification ,medicine.disease ,Infectious Diseases ,Pharmaceutical Preparations ,chemistry ,Pharmacodynamics ,Models, Animal ,Linezolid ,business - Abstract
Preclinical animal models of infection are employed to develop new agents but also to screen among molecules to rank them. There are often major differences between human pharmacokinetic (PK) profiles and those developed by animal models of infection, and these may lead to substantial differences in efficacy relative to that seen in humans. Linezolid is a repurposed agent employed to great effect for therapy of Mycobacterium tuberculosis. In this study, we used the hollow-fiber infection model (HFIM) to evaluate the impact of different pharmacokinetic profiles of mice and nonhuman primates (NHP) versus humans on bacterial cell kill as well as resistance suppression. We examined both plasma and epithelial lining fluid (ELF) profiles. We examined simulated exposures equivalent to 600 mg and 900 mg daily of linezolid in humans. For both plasma and ELF exposures, the murine PK profile provided estimates of effect that were biased low relative to human and NHP PK profiles. Mathematical modeling identified a linkage between minimum concentrations (C(min)) and bacterial kill and peak concentrations (C(peak)) and resistance suppression, with the latter being supported by a prospective validation study. Finding new agents with novel mechanisms of action against M. tuberculosis is difficult. It would be a tragedy to discard a new agent because of a biased estimate of effect in a preclinical animal system. The HFIM provides a system to benchmark evaluation of new compounds in preclinical animal model systems against human PK effects (species scale-up estimates of PK), to safeguard against unwarranted rejection of promising new agents.
- Published
- 2020
15. Building Optimal Three-Drug Combination Chemotherapy Regimens
- Author
-
Walter M. Yamada, Jocelyn Nole, Nino Mtchedlidze, George L. Drusano, Michael Neely, Sarah Kim, Arnold Louie, Stephan Schmidt, Charles A. Peloquin, and Brandon Duncanson
- Subjects
Oncology ,medicine.medical_specialty ,Tuberculosis ,Combination therapy ,Antitubercular Agents ,Leprostatic Agents ,03 medical and health sciences ,chemistry.chemical_compound ,Moxifloxacin ,Internal medicine ,medicine ,Pharmacology (medical) ,Experimental Therapeutics ,030304 developmental biology ,Pharmacology ,0303 health sciences ,030306 microbiology ,business.industry ,Combination chemotherapy ,Mycobacterium tuberculosis ,medicine.disease ,Confidence interval ,Regimen ,Infectious Diseases ,chemistry ,Pharmaceutical Preparations ,Pretomanid ,Drug Therapy, Combination ,Bedaquiline ,business ,medicine.drug - Abstract
Multidrug therapy is often required. Examples include antiviral therapy, nosocomial infections, and, most commonly, anti-Mycobacterium tuberculosis therapy. Our laboratory previously identified a mathematical approach to identify 2-drug regimens with a synergistic or additive interaction using a full factorial study design. Our objective here was to generate a method to identify an optimal 3-drug therapy. We studied M. tuberculosis isolate H37Rv in log-phase growth in flasks. Pretomanid and moxifloxacin were chosen as the base 2-drug regimen. Bedaquiline (plus M2 metabolite) was chosen as the third drug for evaluation. Total bacterial burden and bacterial burden less-susceptible to study drugs were enumerated. A large mathematical model was fit to all the data. This allowed extension to evaluation of the 3-drug regimen by employing a Monte Carlo simulation. Pretomanid plus moxifloxacin demonstrated excellent bacterial kill and suppressed amplification of less-susceptible pathogens. Total bacterial burden was driven to extinction in 3 weeks in 6 of 9 combination therapy evaluations. Only the lowest pretomanid/moxifloxacin exposures in combination did not extinguish the bacterial burden. No combination regimen allowed resistance amplification. Generation of 95% credible intervals about estimates of the interaction parameters α (α(s), α(r-p), and α(r-m)) by bootstrapping showed the interaction was near synergistic. The addition of bedaquiline/M2 metabolite was evaluated by forming a 95% confidence interval regarding the decline in bacterial burden. The addition of bedaquiline/M2 metabolite shortened the time to eradication by 1 week and was significantly different. A model-based system approach to evaluating combinations of 3 agents shows promise to rapidly identify the most promising combinations that can then be trialed.
- Published
- 2020
16. Pharmacokinetics of tedizolid, sutezolid, and sutezolid-M1 in non-human primates
- Author
-
Arnold Louie, JoAnne L. Flynn, Chelsea M Causgrove, Véronique Dartois, George L. Drusano, Charles A. Scanga, Matthew D. Zimmerman, Carolina de Miranda Silva, Stephan Schmidt, Emily Graham, Charles A. Peloquin, Brianne Stein, and Sarah Kim
- Subjects
Primates ,Metabolite ,Cmax ,Pharmaceutical Science ,Tetrazoles ,02 engineering and technology ,Microbial Sensitivity Tests ,Pharmacology ,030226 pharmacology & pharmacy ,Mycobacterium tuberculosis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Medicine ,Animals ,Oxazoles ,Oxazolidinones ,Volume of distribution ,biology ,business.industry ,Prodrug ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Organophosphates ,Anti-Bacterial Agents ,chemistry ,Pharmacodynamics ,Tedizolid ,0210 nano-technology ,business - Abstract
Non-human primates (NHP) are thought to be a good preclinical animal model for tuberculosis because they develop disease characteristics that are similar to humans. The objective of the current study was to determine if NHPs can also be used to reliably predict the exposure of tedizolid, sutezolid, and its biologically active metabolite sutezolid-M1 in humans. The prodrug tedizolid phosphate and sutezolid were administered orally to NHPs either once or twice daily for up to eight days. The active moieties, tedizolid, and sutezolid showed linear pharmacokinetics and respective concentration-time profiles could be described by one-compartment body models with first-order elimination. One additional metabolite compartment with first-order elimination was found appropriate to capture the pharmacokinetics of sutezolid-M1. Once allometrically scaled to humans with a fixed exponent of 0.75 for apparent clearance and 1 for apparent volume of distribution, the AUCs of tedizolid and sutezolid were predicted reasonably well, whereas Cmax was under-predicted for sutezolid. Both NHP and humanized concentration-time profiles will now be used in vitro hollow-fiber pharmacodynamic experiments to determine if differences in drug exposures result in differences in Mycobacterium tuberculosis kill and emergence of resistance.
- Published
- 2020
17. Novel Cassette Assay To Quantify the Outer Membrane Permeability of Five β-Lactams Simultaneously in Carbapenem-Resistant
- Author
-
Tae Hwan, Kim, Xun, Tao, Bartolome, Moya, Yuanyuan, Jiao, Kari B, Basso, Jieqiang, Zhou, Yinzhi, Lang, Dhruvitkumar S, Sutaria, Alexandre P, Zavascki, Afonso L, Barth, Stephanie M, Reeve, Herbert P, Schweizer, Deanna, Deveson Lucas, John D, Boyce, Robert A, Bonomo, Richard E, Lee, Beom Soo, Shin, Arnold, Louie, George L, Drusano, and Jürgen B, Bulitta
- Subjects
monobactams ,Cell Membrane Permeability ,beta-lactams ,carbapenem resistance ,cassette assay ,Microbial Sensitivity Tests ,biochemical phenomena, metabolism, and nutrition ,Therapeutics and Prevention ,outer membrane ,Anti-Bacterial Agents ,Klebsiella pneumoniae ,Bacterial Outer Membrane ,Carbapenem-Resistant Enterobacteriaceae ,Carbapenems ,Enterobacter cloacae ,cephalosporins ,polycyclic compounds ,polymyxin resistance ,LC-MS/MS ,permeability ,Research Article - Abstract
Antimicrobial resistance is causing a global human health crisis and is affecting all antibiotic classes. While β-lactams have been commonly used against susceptible isolates of Klebsiella pneumoniae and Enterobacter cloacae, carbapenem-resistant isolates are spreading worldwide and pose substantial clinical challenges. Rapid penetration of β-lactams leads to high drug concentrations at their periplasmic target sites, allowing β-lactams to more completely inactivate their target receptors. Despite this, there are limited tangible data on the permeability of β-lactams through the outer membranes of many Gram-negative pathogens. This study presents a novel, cassette assay, which can simultaneously characterize the permeability of five β-lactams in multidrug-resistant clinical isolates. We show that carbapenems, and especially imipenem, penetrate the outer membrane of K. pneumoniae and E. cloacae substantially faster than noncarbapenem β-lactams. The ability to efficiently characterize the outer membrane permeability is critical to optimize the use of β-lactams and combat carbapenem-resistant isolates., Poor penetration through the outer membrane (OM) of Gram-negative bacteria is a major barrier of antibiotic development. While β-lactam antibiotics are commonly used against Klebsiella pneumoniae and Enterobacter cloacae, there are limited data on OM permeability especially in K. pneumoniae. Here, we developed a novel cassette assay, which can simultaneously quantify the OM permeability to five β-lactams in carbapenem-resistant K. pneumoniae and E. cloacae. Both clinical isolates harbored a blaKPC-2 and several other β-lactamases. The OM permeability of each antibiotic was studied separately (“discrete assay”) and simultaneously (“cassette assay”) by determining the degradation of extracellular β-lactam concentrations via multiplex liquid chromatography-tandem mass spectrometry analyses. Our K. pneumoniae isolate was polymyxin resistant, whereas the E. cloacae was polymyxin susceptible. Imipenem penetrated the OM at least 7-fold faster than meropenem for both isolates. Imipenem penetrated E. cloacae at least 258-fold faster and K. pneumoniae 150-fold faster compared to aztreonam, cefepime, and ceftazidime. For our β-lactams, OM permeability was substantially higher in the E. cloacae compared to the K. pneumoniae isolate (except for aztreonam). This correlated with a higher OmpC porin production in E. cloacae, as determined by proteomics. The cassette and discrete assays showed comparable results, suggesting limited or no competition during influx through OM porins. This cassette assay allowed us, for the first time, to efficiently quantify the OM permeability of multiple β-lactams in carbapenem-resistant K. pneumoniae and E. cloacae. Characterizing the OM permeability presents a critical contribution to combating the antimicrobial resistance crisis and enables us to rationally optimize the use of β-lactam antibiotics.
- Published
- 2020
18. GC-072: A Novel Therapeutic Candidate for Oral Treatment of Melioidosis and Infections Caused by Select Biothreat Pathogens
- Author
-
Herbert P. Schweizer, Kelly L. Warfield, Sunisa Chirakul, Jeffry D Shearer, Michelle L Saylor, Henry S. Heine, George L. Drusano, Steven D. Zumbrun, Anthony M. Treston, Christine M Butler, and Arnold Louie
- Subjects
Melioidosis ,medicine.drug_class ,Burkholderia ,Antibiotics ,Ceftazidime ,Microbiology ,03 medical and health sciences ,Burkholderia mallei ,antibiotic ,Medicine ,Pharmacology (medical) ,Experimental Therapeutics ,Pathogen ,Francisella tularensis ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,biodefense ,030306 microbiology ,business.industry ,Burkholderia pseudomallei ,GC-072 ,biology.organism_classification ,medicine.disease ,bacterial infections and mycoses ,Bacillus anthracis ,Infectious Diseases ,bacteria ,melioidosis ,business ,medicine.drug - Abstract
Burkholderia pseudomallei, the etiological agent of melioidosis, is a Gram-negative bacterium with additional concern as a biothreat pathogen. The mortality rate from B. pseudomallei varies depending on the type of infection and extent of available health care; in the case of septicemia, left untreated, it can range from 50% to 90%. Current therapy for melioidosis is biphasic, consisting of parenteral acute-phase treatment for 2 weeks or longer, followed by oral eradication-phase treatment lasting several months., Burkholderia pseudomallei, the etiological agent of melioidosis, is a Gram-negative bacterium with additional concern as a biothreat pathogen. The mortality rate from B. pseudomallei varies depending on the type of infection and extent of available health care; in the case of septicemia, left untreated, it can range from 50% to 90%. Current therapy for melioidosis is biphasic, consisting of parenteral acute-phase treatment for 2 weeks or longer, followed by oral eradication-phase treatment lasting several months. An effective oral therapeutic for outpatient treatment of acute-phase melioidosis is needed. GC-072 is a potent, 4-oxoquinolizine antibiotic with selective inhibitory activity against bacterial topoisomerases. GC-072 has demonstrated in vitro potency against susceptible and drug-resistant strains of B. pseudomallei and is also active against Burkholderia mallei, Bacillus anthracis, Yersinia pestis, and Francisella tularensis. GC-072 is bactericidal both extra- and intracellularly, with rapid killing noted within a few hours and reduced development of resistance compared to that for ceftazidime. GC-072, delivered intragastrically to mimic oral administration, promoted dose-dependent survival in mice using lethal inhalational models of B. pseudomallei infection following exposure to a 24- or 339-LD50 (50% lethal dose) challenge with B. pseudomallei strain 1026b. Overall, GC-072 appears to be a strong candidate for first-line oral treatment of melioidosis.
- Published
- 2019
19. Pharmacodynamics of Ceftazidime plus Avibactam against KPC-2-Bearing Isolates of Klebsiella pneumoniae in a Hollow Fiber Infection Model
- Author
-
M. Hong Nguyen, Nino Mtchedlidze, Cornelius J. Clancy, Ryan K. Shields, Arnold Louie, George L. Drusano, and Michael Vicciarelli
- Subjects
Pharmacology ,0303 health sciences ,030306 microbiology ,Kill rate ,medicine.drug_class ,Klebsiella pneumoniae ,Avibactam ,Polymyxin ,Ceftazidime ,Liter ,Biology ,biology.organism_classification ,Microbiology ,03 medical and health sciences ,Regimen ,chemistry.chemical_compound ,0302 clinical medicine ,Infectious Diseases ,chemistry ,Pharmacodynamics ,medicine ,Pharmacology (medical) ,030212 general & internal medicine ,medicine.drug - Abstract
Ceftazidime-avibactam (CAZ/AVI) combines ceftazidime with a diazabicyclooctane non-β-lactam β-lactamase inhibitor. This has potent inhibitory activity against KPC-type enzymes. We studied activity of clinically relevant regimens of CAZ/AVI against two KPC-2-bearing Klebsiella pneumoniae isolates (sequence type 258 recovered sequentially from the same patient) with and without ompK36 mutations in a hollow fiber infection model. The baseline total bacterial burden exceeded 109 CFU. For both isolates, there was early multi-log CFU/ml reductions in the bacterial burden for all regimens. Bacterial subpopulations with reduced susceptibilities to CAZ/AVI were isolated only from the no-treatment control arms. All CAZ/AVI regimens resulted in undetectable colony counts between days 6 and 8. At day 10, the total volume of each CAZ/AVI arm was plated, with no organisms recovered from any regimen, documenting complete eradication. A population model was fit to avibactam concentrations and total colony count outputs. The model fit was acceptable and demonstrated a large kill rate constant (K kill = 6.29 h-1) and a relatively low avibactam concentration at which kill rate was half maximal (C 50 = 2.19 mg/liter), concordant with the observed bacterial burden decline. A threshold analysis identified time > 4 mg/liter of avibactam as the index most closely linked to bacterial burden decline. Given the clinical outcomes seen with KPC-bearing organisms and the toxicities that occur when patients are treated with currently available polymyxins, drugs such as CAZ/AVI should have a prominent place in early therapy.
- Published
- 2019
20. Comparable Efficacy and Better Safety of Double β-Lactam Combination Therapy versus β‑Lactam plus Aminoglycoside in Gram-Negative Bacteria in Randomized, Controlled Trials
- Author
-
Dhruvitkumar S. Sutaria, Bartolome Moya, Hsin-Yin Tsai, Jürgen B. Bulitta, John D. Boyce, Mong-Jen Chen, Tae Hwan Kim, Yuanyuan Jiao, Alexandre P. Zavascki, Arnold Louie, Brian T. Tsuji, Robert A. Bonomo, Deanna Deveson Lucas, Xun Tao, and George L. Drusano
- Subjects
medicine.medical_specialty ,Combination therapy ,Clinical Therapeutics ,beta-Lactams ,law.invention ,03 medical and health sciences ,Ototoxicity ,Randomized controlled trial ,law ,Internal medicine ,Humans ,Medicine ,Pharmacology (medical) ,Adverse effect ,Randomized Controlled Trials as Topic ,030304 developmental biology ,Pharmacology ,0303 health sciences ,030306 microbiology ,business.industry ,Aminoglycoside ,medicine.disease ,Anti-Bacterial Agents ,Clinical trial ,Aminoglycosides ,Treatment Outcome ,Infectious Diseases ,Meta-analysis ,Relative risk ,Tobramycin ,Drug Therapy, Combination ,Gram-Negative Bacterial Infections ,business - Abstract
There is a great need for efficacious therapies against Gram-negative bacteria. Double β-lactam combination(s) (DBL) are relatively safe, and preclinical data are promising; however, their clinical role has not been well defined. We conducted a metaanalysis of the clinical and microbiological efficacy of DBL compared to β-lactam plus aminoglycoside combinations (BLAG). PubMed, Embase, ISI Web of Knowledge, and Cochrane Controlled Trials Register database were searched through July 2018. We included randomized controlled clinical trials that compared DBL with BLAG combinations. Clinical response was used as the primary outcome and microbiological response in Gram-negative bacteria as the secondary outcome; sensitivity analyses were performed for Pseudomonas aeruginosa, Klebsiella spp., and Escherichia coli. Heterogeneity and risk of bias were assessed. Safety results were classified by systems and organs. Thirteen studies evaluated 2,771 cases for clinical response and 665 cases for microbiological response in various Gram-negative species. DBL achieved slightly, but not significantly, better clinical response (risk ratio, 1.05; 95% confidence interval [CI], 0.99 to 1.11) and microbiological response in Gram-negatives (risk ratio, 1.11; 95% CI, 0.99 to 1.25) compared with BLAG. Sensitivity analyses by pathogen showed the same trend. No significant heterogeneity across studies was found. DBL was significantly safer than BLAG regarding renal toxicity (6.6% versus 8.8%, P = 0.0338) and ototoxicity (0.7 versus 3.1%, P = 0.0137). Other adverse events were largely comparable. Overall, empirically designed DBL showed comparable clinical and microbiological responses across different Gram-negative species, and were significantly safer than BLAG. Therefore, DBL should be rationally optimized via the latest translational approaches, leveraging mechanistic insights and newer β-lactams for future evaluation in clinical trials.
- Published
- 2019
21. Pharmacodynamics of Ceftazidime plus Avibactam against KPC-2-Bearing Isolates of
- Author
-
G L, Drusano, Ryan K, Shields, Nino, Mtchedlidze, M Hong, Nguyen, Cornelius J, Clancy, Michael, Vicciarelli, and Arnold, Louie
- Subjects
Drug Combinations ,Klebsiella pneumoniae ,Carbapenem-Resistant Enterobacteriaceae ,Bacterial Proteins ,Humans ,Experimental Therapeutics ,Microbial Sensitivity Tests ,Azabicyclo Compounds ,Ceftazidime ,beta-Lactamases ,Anti-Bacterial Agents ,Klebsiella Infections - Abstract
Ceftazidime-avibactam (CAZ/AVI) combines ceftazidime with a diazabicyclooctane non-β-lactam β-lactamase inhibitor. This has potent inhibitory activity against KPC-type enzymes. We studied activity of clinically relevant regimens of CAZ/AVI against two KPC-2-bearing Klebsiella pneumoniae isolates (sequence type 258 recovered sequentially from the same patient) with and without ompK36 mutations in a hollow fiber infection model. The baseline total bacterial burden exceeded 10(9) CFU. For both isolates, there was early multi-log CFU/ml reductions in the bacterial burden for all regimens. Bacterial subpopulations with reduced susceptibilities to CAZ/AVI were isolated only from the no-treatment control arms. All CAZ/AVI regimens resulted in undetectable colony counts between days 6 and 8. At day 10, the total volume of each CAZ/AVI arm was plated, with no organisms recovered from any regimen, documenting complete eradication. A population model was fit to avibactam concentrations and total colony count outputs. The model fit was acceptable and demonstrated a large kill rate constant (K(kill) = 6.29 h(−1)) and a relatively low avibactam concentration at which kill rate was half maximal (C(50) = 2.19 mg/liter), concordant with the observed bacterial burden decline. A threshold analysis identified time > 4 mg/liter of avibactam as the index most closely linked to bacterial burden decline. Given the clinical outcomes seen with KPC-bearing organisms and the toxicities that occur when patients are treated with currently available polymyxins, drugs such as CAZ/AVI should have a prominent place in early therapy.
- Published
- 2019
22. Effect of Moxifloxacin plus Pretomanid against Mycobacterium tuberculosis in Log Phase, Acid Phase, and Nonreplicating-Persister Phase in an In Vitro Assay
- Author
-
Jocelyn Nole, Amirhossein Hajihosseini, Carolina de Miranda Silva, Arnold Louie, Jenny Myrick, George L. Drusano, and Stephan Schmidt
- Subjects
Pharmacology ,0303 health sciences ,Tuberculosis ,Combination therapy ,biology ,030306 microbiology ,Chemistry ,Bacterial growth ,biology.organism_classification ,medicine.disease ,In vitro ,Mycobacterium tuberculosis ,03 medical and health sciences ,chemistry.chemical_compound ,Infectious Diseases ,Moxifloxacin ,Phase (matter) ,Pretomanid ,medicine ,Pharmacology (medical) ,medicine.drug - Abstract
Combination therapy is a successful approach to treat tuberculosis in patients with susceptible strains of Mycobacterium tuberculosis However, the emergence of resistant strains requires identification of new, effective therapies. Pretomanid (PA824) and moxifloxacin (MXF) are promising options currently under evaluation in clinical trials for the treatment of susceptible and resistant mycobacteria. We applied our recently described screening strategy to characterize the interaction between PA824 and MXF toward the killing of M. tuberculosis in logarithmic growth phase (log phase), acid phase, and nonreplicating-persister (NRP) phase. Respective in vitro data generated for the H37Rv and 18b strains were evaluated in a microdilution plate system containing both drugs in combination. The Universal Response Surface Approach model from Greco et al. (W. R. Greco, G. Bravo, and J. C. Parsons, Pharmacol Rev 47:331-385, 1995) was used to characterize the nature of the interaction between both drugs; synergistic or additive combinations would prompt additional evaluation in the hollow-fiber infection model (HFIM) and in animal studies. The interaction between MXF and PA824 was additive against M. tuberculosis organisms in acid phase (interaction parameter [α] = 5.56e-8 [95% confidence interval {CI} = -0.278 to 0.278] and α = 0.408 [95% CI = 0.105 to 0.711], respectively), NRP phase (α = 0.625 [95% CI = -0.556 to 1.81] and α = 2.92 [95% CI = 0.215 to 5.63], respectively), and log phase (α = 1.57e-6 [95% CI = -0.930 to 0.930] and α = 1.83e-6 [95% CI = -0.929 and 0.929], respectively), prompting further testing of this promising combination for the treatment of tuberculosis in the HFIM and in animal studies.
- Published
- 2019
23. Activity of Moxifloxacin against Mycobacterium tuberculosis in Acid Phase and Nonreplicative-Persister Phenotype Phase in a Hollow-Fiber Infection Model
- Author
-
Brandon Duncanson, Jocelyn Nole, Charles A. Peloquin, David Brown, Stephan Schmidt, Charles A. Scanga, Jenny Myrick, Michael Maynard, Arnold Louie, Michael Neely, and George L. Drusano
- Subjects
0301 basic medicine ,Tuberculosis ,medicine.drug_class ,Moxifloxacin ,030106 microbiology ,Population ,Antibiotics ,Antitubercular Agents ,Colony Count, Microbial ,Microbial Sensitivity Tests ,Biology ,Models, Biological ,Microbiology ,Mycobacterium tuberculosis ,03 medical and health sciences ,Drug Resistance, Bacterial ,medicine ,Humans ,Experimental Therapeutics ,Drug Dosage Calculations ,Pharmacology (medical) ,education ,Pharmacology ,education.field_of_study ,Models, Statistical ,Dose-Response Relationship, Drug ,medicine.disease ,biology.organism_classification ,Culture Media ,Phenotype ,030104 developmental biology ,Infectious Diseases ,Streptomycin ,Pharmacodynamics ,Diffusion Chambers, Culture ,Metabolic Networks and Pathways ,Bacteria ,medicine.drug - Abstract
A major goal for improving tuberculosis therapy is to identify drug regimens with improved efficacy and shorter treatment durations. Shorter therapies improve patient adherence to the antibiotic regimens that, in turn, decreases resistance emergence. M. tuberculosis exists in multiple metabolic states. At the initiation of therapy, the bulk of the population is in Log-Phase growth. Consequently, it is logical to focus initial therapy on these organisms. Moxifloxacin has good early bactericidal activity against Log-Phase growth bacteria and is a logical component of initial therapy. It would be optimal if this agent also possessed activity against Acid-Phase and Non-Replicative Persister (NRP) Phenotype organisms. We studied multiple exposures to moxifloxacin (equivalent to 200 mg-800 mg daily) in our Hollow Fiber Infection Model against strain H37Rv in Acid-Phase and against strain 18b in streptomycin starvation, which is a model for NRP-Phase organisms. Moxifloxacin possesses good activity against Acid-Phase organisms, generating from 3.75 Log 10 (CFU/ml) cell kill (200 mg daily) to 5.16 Log 10 (CFU/ml) cell kill (800 mg daily) over the 28 days of the experiment. Moxifloxacin also has activity against the streptomycin-starved strain 18b. The 400 to 800 mg daily regimens achieved extinction at day 28, while the no-treatment control still had 1.96 Log 10 (CFU/ml) culturable. The lowest dose (200 mg daily) still had 0.7 Log 10 (CFU/ml) measurable at day 28, a net kill of 1.26 Log 10 (CFU/ml). Moxifloxacin is an attractive agent for early therapy, as it possesses activity against three metabolic states of M. tuberculosis .
- Published
- 2018
24. Natural History of Francisella tularensis in Aerosol-Challenged BALB/c Mice
- Author
-
George L. Drusano, Robert T. Cass, Ryan Cirz, Henry S. Heine, Lara Chuvala, Arnold Louie, and Renaldo Riggins
- Subjects
0301 basic medicine ,030106 microbiology ,Bacteremia ,Spleen ,BALB/c ,Pathogenesis ,Tularemia ,Mice ,03 medical and health sciences ,medicine ,Animals ,Experimental Therapeutics ,Pharmacology (medical) ,Francisella tularensis ,Lung ,Aerosols ,Pharmacology ,Mice, Inbred BALB C ,biology ,Models, Theoretical ,biology.organism_classification ,medicine.disease ,Bacterial Load ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Splenic Tissue ,Immunology ,Disease Progression ,Female - Abstract
The objective of this study was to evaluate the natural history and pathogenesis of Francisella tularensis in a murine model of inhalational tularemia with the SchuS4 strain. Before the efficacy of antimicrobials could be assessed in this model, further model development was required to determine the optimal time to start therapy. This study helped define the time course of infection after aerosol challenge by quantifying the presence of bacteria in lung, blood, and spleen at multiple harvest points. In this study, mice were infected via a targeted inhaled dose of 100 50% lethal doses (LD 50 s) (LD 50 = 300 CFU) of F. tularensis by whole-body aerosol. At 1, 24, 36, 48, 60, 72, 75, 78, 81, 84, 87, and 90 h postchallenge, groups of 15 animals were sacrificed and blood, lung, and splenic tissue samples were harvested, homogenized, plated, and incubated to evaluate the bacterial load in those tissues. It was determined that of the 3 sample types harvested, splenic tissue provided the most consistent bacterial counts, which steadily increased with the progressing infection. Further, it was determined that lung samples from all (15/15) animals were positive for infection at 75 h postaerosolization and that 14/15 animals had positive splenic tissue counts. Bacterial levels in blood were not predictive of treatment initiation. For future therapeutic evaluation studies in this model using F. tularensis (SchuS4), it was determined that therapy should be initiated at 75 h postchallenge and validated by spleen involvement.
- Published
- 2016
25. Breakpoint determination when multiple organisms are tested for effect targets
- Author
-
Arnold Louie and George L. Drusano
- Subjects
Percentile ,Mice, Inbred BALB C ,Breakpoint ,Pharmaceutical Science ,02 engineering and technology ,Computational biology ,Microbial Sensitivity Tests ,Biology ,021001 nanoscience & nanotechnology ,030226 pharmacology & pharmacy ,Anti-Bacterial Agents ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Drug Delivery Systems ,Enterobacteriaceae ,Murine model ,Target attainment ,Animals ,Humans ,Female ,0210 nano-technology ,Monte Carlo Method - Abstract
Determination of the susceptibility breakpoint for antibiotics is important, as it guides the use of agents in the clinical setting. Currently, breakpoints are often evaluated using a Probability of Target Attainment Analysis in which the targets are set through pre-clinical experiments, often by examining a strain of a target pathogen in a murine model such as a neutropenic thigh infection model. However, regulatory authorities are often rightly concerned about the setting of breakpoints when a number of isolates of target pathogens are evaluated and there is a sizeable spread of the drug exposures necessary to achieve the target with a sufficiently high (usually 90%) probability. Here, we propose a method for supporting a breakpoint determination for this circumstance. We examined 8 isolates of resistant Enterobacteriaceae in a neutropenic murine thigh infection model. The stasis exposure was determined and ranged from 5.70 to 43.5 AUC/MIC Ratio. The mean ± standard deviation was 20.05 ± 13.05. A 5000-iterate Monte Carlo simulation was performed to generate a range of stasis targets and Probability of Target Attainment Analyses were calculated at the 1st, 5th, 10th, 25th, 50th, 75th, 90th, 95th, and 99th percentiles of the distribution. Breakpoints were determined at each percentile. Breakpoints ranged from 2 mg/L to 32 mg/L. A weighted (by the percentages of the distribution) breakpoint was calculated and determined to be 4 mg/L. This method is a rational approach to identifying breakpoints when there is substantial between-isolate variability in exposure targets.
- Published
- 2018
26. The Combination of Fosfomycin plus Meropenem Is Synergistic for Pseudomonas aeruginosa PAO1 in a Hollow-Fiber Infection Model
- Author
-
Michael Vicchiarelli, Michael Neely, Walter M. Yamada, David Brown, Brandon Duncanson, Arnold Louie, George L. Drusano, and Michael Maynard
- Subjects
0301 basic medicine ,Combination therapy ,medicine.medical_treatment ,030106 microbiology ,Colony Count, Microbial ,Microbial Sensitivity Tests ,Pharmacology ,Fosfomycin ,medicine.disease_cause ,Meropenem ,Models, Biological ,03 medical and health sciences ,Drug Resistance, Bacterial ,medicine ,Humans ,Pharmacology (medical) ,Drug Dosage Calculations ,Pseudomonas Infections ,Experimental Therapeutics ,Pathogen ,Dose-Response Relationship, Drug ,business.industry ,Pseudomonas aeruginosa ,Combination chemotherapy ,Drug Synergism ,biochemical phenomena, metabolism, and nutrition ,Anti-Bacterial Agents ,Culture Media ,Drug Combinations ,Infectious Diseases ,Phenotype ,Intravenous therapy ,Pharmacodynamics ,Diffusion Chambers, Culture ,business ,Factor Analysis, Statistical ,Metabolic Networks and Pathways ,medicine.drug - Abstract
Treating high-density bacterial infections is a challenging clinical problem. We have a paucity of new agents that can address this problem. Pseudomonas aeruginosa is a particularly difficult pathogen to treat effectively because of the plethora of resistance mechanisms it carries. Fosfomycin is an agent discovered circa 40 years ago. Recently, it has been resurrected in the United States and studied for intravenous therapy. We hypothesized that, to maximize its utility, it would require combination chemotherapy when used in a clinical circumstance in high-bacterial-burden infections. We chose to examine the combination of meropenem plus fosfomycin. These agents were studied in the hollow-fiber infection model. We utilized a fully factorial study design, looking at 2 doses of meropenem alone (1 and 2 g 8-hourly) and two doses of fosfomycin alone (6 and 8 g 8-hourly), as well as all possible combinations plus a no-treatment control. We used a high-dimensional model of 5 inhomogeneous differential equations with 5 system outputs to analyze all data simultaneously. Combination therapy outperformed all monotherapy regimens, with all combinations driving >6 log(10) CFU/ml of bacterial killing. Combination therapy was able to counterselect resistance emergence (meropenem mutants being killed by the combination, as well as fosfomycin mutants being killed by the combination) in all regimens studied. The analysis demonstrated that the combination was significantly synergistic for bacterial cell killing and resistance suppression. Meropenem plus fosfomycin is a promising combination for therapy of high-burden Pseudomonas aeruginosa infections and requires further study.
- Published
- 2018
27. Effect of Moxifloxacin plus Pretomanid against
- Author
-
Carolina, de Miranda Silva, Amirhossein, Hajihosseini, Jenny, Myrick, Jocelyn, Nole, Arnold, Louie, Stephan, Schmidt, and George L, Drusano
- Subjects
Drug Combinations ,Models, Statistical ,Drug Resistance, Fungal ,Nitroimidazoles ,Moxifloxacin ,Antitubercular Agents ,Drug Interactions ,Experimental Therapeutics ,Microbial Sensitivity Tests ,Mycobacterium tuberculosis - Abstract
Combination therapy is a successful approach to treat tuberculosis in patients with susceptible strains of Mycobacterium tuberculosis. However, the emergence of resistant strains requires identification of new, effective therapies. Pretomanid (PA824) and moxifloxacin (MXF) are promising options currently under evaluation in clinical trials for the treatment of susceptible and resistant mycobacteria. We applied our recently described screening strategy to characterize the interaction between PA824 and MXF toward the killing of M. tuberculosis in logarithmic growth phase (log phase), acid phase, and nonreplicating-persister (NRP) phase. Respective in vitro data generated for the H37Rv and 18b strains were evaluated in a microdilution plate system containing both drugs in combination. The Universal Response Surface Approach model from Greco et al. (W. R. Greco, G. Bravo, and J. C. Parsons, Pharmacol Rev 47:331–385, 1995) was used to characterize the nature of the interaction between both drugs; synergistic or additive combinations would prompt additional evaluation in the hollow-fiber infection model (HFIM) and in animal studies. The interaction between MXF and PA824 was additive against M. tuberculosis organisms in acid phase (interaction parameter [α] = 5.56e−8 [95% confidence interval {CI} = −0.278 to 0.278] and α = 0.408 [95% CI = 0.105 to 0.711], respectively), NRP phase (α = 0.625 [95% CI = −0.556 to 1.81] and α = 2.92 [95% CI = 0.215 to 5.63], respectively), and log phase (α = 1.57e−6 [95% CI = −0.930 to 0.930] and α = 1.83e−6 [95% CI = −0.929 and 0.929], respectively), prompting further testing of this promising combination for the treatment of tuberculosis in the HFIM and in animal studies.
- Published
- 2018
28. Four Decades of β-Lactam Antibiotic Pharmacokinetics in Cystic Fibrosis
- Author
-
Bartolome Moya, Alexandre P. Zavascki, Beom Soo Shin, Stefanie K. Drescher, Jürgen B. Bulitta, Antonio Oliver, Yuanyuan Jiao, Fritz Sörgel, Arnold Louie, Cornelia B. Landersdorfer, Brian T. Tsuji, Mathias Wittau, Xun Tao, and George L. Drusano
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cystic Fibrosis ,medicine.drug_class ,030106 microbiology ,Population ,Antibiotics ,Medizin ,Body size ,beta-Lactams ,Cystic fibrosis ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Healthy volunteers ,medicine ,Humans ,Pharmacology (medical) ,030212 general & internal medicine ,education ,Pharmacology ,Volume of distribution ,education.field_of_study ,business.industry ,medicine.disease ,Anti-Bacterial Agents ,Lean body mass ,business - Abstract
The pharmacokinetics (PK) of β-lactam antibiotics in cystic fibrosis (CF) patients has been compared with that in healthy volunteers for over four decades; however, no quantitative models exist that explain the PK differences between CF patients and healthy volunteers in older and newer studies. Our aims were to critically evaluate these studies and explain the PK differences between CF patients and healthy volunteers. We reviewed all 16 studies that compared the PK of β-lactams between CF patients and healthy volunteers within the same study. Analysis of covariance (ANCOVA) models were developed. In four early studies that compared adolescent, lean CF patients with adult healthy volunteers, clearance (CL) in CF divided by that in healthy volunteers was 1.72 ± 0.90 (average ± standard deviation); in four additional studies comparing age-matched (primarily adult) CF patients with healthy volunteers, this ratio was 1.46 ± 0.16. The CL ratio was 1.15 ± 0.11 in all eight studies that compared CF patients and healthy volunteers who were matched in age, body size and body composition, or that employed allometric scaling by lean body mass (LBM). Volume of distribution was similar between subject groups after scaling by body size. For highly protein-bound β-lactams, the unbound fraction was up to 2.07-fold higher in older studies that compared presumably sicker CF patients with healthy volunteers. These protein-binding differences explained over half of the variance for the CL ratio (p
- Published
- 2018
29. First Penicillin-Binding Protein Occupancy Patterns of β-Lactams and β-Lactamase Inhibitors in Klebsiella pneumoniae
- Author
-
Bartolome Moya, Jürgen B. Bulitta, Arnold Louie, Kari B. Green, Yuanyuan Jiao, Dhruvitkumar S. Sutaria, Xun Tao, Tae Hwan Kim, and George L. Drusano
- Subjects
0301 basic medicine ,Penicillin binding proteins ,Klebsiella pneumoniae ,Avibactam ,030106 microbiology ,Ceftazidime ,Microbial Sensitivity Tests ,Aztreonam ,beta-Lactams ,03 medical and health sciences ,chemistry.chemical_compound ,Bacterial Proteins ,polycyclic compounds ,medicine ,Penicillin-Binding Proteins ,Pharmacology (medical) ,Mecillinam ,Beta-Lactamase Inhibitors ,Pharmacology ,Principal Component Analysis ,biology ,Chemistry ,Amdinocillin ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Enterobacteriaceae ,030104 developmental biology ,Infectious Diseases ,Carbapenems ,Biochemistry ,beta-Lactamase Inhibitors ,medicine.drug - Abstract
Penicillin-binding proteins (PBPs) are the high-affinity target sites of all β-lactam antibiotics in bacteria. It is well known that each β-lactam covalently binds to and thereby inactivates different PBPs with various affinities. Despite β-lactams serving as the cornerstone of our therapeutic armamentarium against Klebsiella pneumoniae , PBP binding data are missing for this pathogen. We aimed to generate the first PBP binding data on 13 chemically diverse and clinically relevant β-lactams and β-lactamase inhibitors in K. pneumoniae . PBP binding was determined using isolated membrane fractions from K. pneumoniae strains ATCC 43816 and ATCC 13883. Binding reactions were conducted using β-lactam concentrations from 0.0075 to 256 mg/liter (or 128 mg/liter). After β-lactam exposure, unbound PBPs were labeled by Bocillin FL. Binding affinities (50% inhibitory concentrations [IC 50 ]) were reported as the β-lactam concentrations that half-maximally inhibited Bocillin FL binding. PBP occupancy patterns by β-lactams were consistent across both strains. Carbapenems bound to all PBPs, with PBP2 and PBP4 as the highest-affinity targets (IC 50 , 50 , 50 , 2 mg/liter). Aztreonam showed high affinity for PBP3 (IC 50 , 0.06 to 0.12 mg/liter). Ceftazidime bound PBP3 at low concentrations (IC 50 , 0.06 to 0.25 mg/liter) and PBP1a/b at higher concentrations (4 mg/liter), whereas cefepime bound PBPs 1 to 4 at more even concentrations (IC 50 , 0.015 to 2 mg/liter). These PBP binding data on a comprehensive set of 13 clinically relevant β-lactams and β-lactamase inhibitors in K. pneumoniae enable, for the first time, the rational design and optimization of double β-lactam and β-lactam–β-lactamase inhibitor combinations.
- Published
- 2018
30. Dose optimization of moxifloxacin and linezolid against tuberculosis using mathematical modeling and simulation
- Author
-
Charles A. Peloquin, Arnold Louie, Kenneth H. Rand, David L. Brown, George L. Drusano, Michael Maynard, M. Tobias Heinrichs, Sherwin K. B. Sy, and Hartmut Derendorf
- Subjects
0301 basic medicine ,Microbiology (medical) ,Drug ,Tuberculosis ,media_common.quotation_subject ,030106 microbiology ,Population ,Moxifloxacin ,Drug resistance ,Pharmacology ,Mycobacterium tuberculosis ,03 medical and health sciences ,chemistry.chemical_compound ,Minimum inhibitory concentration ,0302 clinical medicine ,Drug Resistance, Bacterial ,Medicine ,Humans ,Pharmacology (medical) ,030212 general & internal medicine ,education ,media_common ,education.field_of_study ,Microbial Viability ,biology ,business.industry ,Linezolid ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Models, Theoretical ,biology.organism_classification ,medicine.disease ,Anti-Bacterial Agents ,Infectious Diseases ,Treatment Outcome ,chemistry ,business ,medicine.drug - Abstract
Introduction There is an urgent need for new anti-tuberculosis (TB) drugs and optimization of current TB treatment. Moxifloxacin and linezolid are valuable options for the treatment of drug-resistant TB; however, it is crucial to find a dose at which these drugs not only show high efficacy but also suppress the development of further drug resistance. Methods Activity of moxifloxacin and linezolid against Mycobacterium tuberculosis was studied in the hollow-fiber infection model system in log-phase growth under neutral pH and slow growth in an acidic environment. Doses that achieved maximum bacterial kill while suppressing the emergence of drug resistance were determined. Through Monte Carlo simulations the quantitative output of this in vitro study was bridged to the human patient population to inform optimal dosage regimens while accounting for clinical minimum inhibitory concentration (MIC) distributions. Results and Discussion Moxifloxacin activity was significantly decreased in an acidified environment. The loss of activity was compensated by accumulation of the drug in TB lung lesions; therefore, moderate efficacy can be expected. Moxifloxacin 800 mg/day is the dose that most likely leads to resistance suppression while exerting maximum bacterial kill. Linezolid demonstrated very good activity even at a reduced pH. Linezolid 900 mg once-daily (QD) is likely to achieve a maximum killing effect and prevent the emergence of drug resistance; 600 mg QD in a robust drug regimen may have similar potential.
- Published
- 2018
31. Effect of Linezolid plus Bedaquiline against Mycobacterium tuberculosis in Log Phase, Acid Phase, and Nonreplicating-Persister Phase in an
- Author
-
Jocelyn Nole, Carolina de Miranda Silva, Jenny Myrick, Amirhossein Hajihosseini, Stephan Schmidt, Arnold Louie, and George L. Drusano
- Subjects
0301 basic medicine ,Drug ,Tuberculosis ,Combination therapy ,media_common.quotation_subject ,030106 microbiology ,Antitubercular Agents ,Drug action ,Microbial Sensitivity Tests ,Pharmacology ,Mycobacterium tuberculosis ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Resistance, Multiple, Bacterial ,Tuberculosis, Multidrug-Resistant ,Medicine ,Pharmacology (medical) ,Experimental Therapeutics ,Diarylquinolines ,media_common ,biology ,business.industry ,Linezolid ,Drug Synergism ,medicine.disease ,biology.organism_classification ,In vitro ,030104 developmental biology ,Infectious Diseases ,chemistry ,Bedaquiline ,business - Abstract
Tuberculosis is the ninth-leading cause of death worldwide. Treatment success is approximately 80% for susceptible strains and decreases to 30% for extensively resistant strains. Shortening the therapy duration for Mycobacterium tuberculosis is a major goal, which can be attained with the use of combination therapy. However, the identification of the most promising combination is a challenge given the quantity of older and newer agents available. Our objective was to identify promising 2-drug combinations using an in vitro strategy to ultimately be tested in an in vitro hollow fiber infection model (HFIM) and in animal models. We studied the effect of the combination of linezolid (LZD) and bedaquiline (BDQ) on M. tuberculosis strain H37Rv in log- and acid-phase growth and M. tuberculosis strain 18b in log- and nonreplicating-persister-phase growth in a plate system containing a 9-by-8 matrix of concentrations of both drugs alone and in combinations. A characterization of the interaction as antagonistic, additive, or synergistic was performed using the Greco universal response surface approach (URSA) model. Our results indicate that the interaction between LZD and BDQ is additive for bacterial killing in both strains for both of the metabolic states tested. This prescreen strategy was suitable to identify LZD and BDQ as a promising combination to be further tested in the HFIM. The presence of nonoverlapping mechanisms of drug action suggests each drug in the combination will likely be effective in suppressing the emergence of resistance by M. tuberculosis to the companion drug, which holds promise in improving treatment outcomes for tuberculosis.
- Published
- 2018
32. Linezolid Kills Acid-Phase and Nonreplicative-Persister-Phase Mycobacterium tuberculosis in a Hollow-Fiber Infection Model
- Author
-
Stephan Schmidt, Jenny Myrick, Charles A. Peloquin, Arnold Louie, Jocelyn Nole, George L. Drusano, Brandon Duncanson, Charles A. Scanga, Michael Neely, David Brown, and Michael Maynard
- Subjects
0301 basic medicine ,Tuberculosis ,030106 microbiology ,Population ,Antitubercular Agents ,Models, Biological ,Drug Administration Schedule ,Microbiology ,Mycobacterium tuberculosis ,03 medical and health sciences ,chemistry.chemical_compound ,Cmin ,medicine ,Therapy duration ,Animals ,Humans ,Pharmacology (medical) ,Drug Dosage Calculations ,Experimental Therapeutics ,Dosing ,education ,Pharmacology ,education.field_of_study ,Models, Statistical ,biology ,business.industry ,Linezolid ,medicine.disease ,Antimicrobial ,biology.organism_classification ,Infectious Diseases ,chemistry ,Area Under Curve ,Diffusion Chambers, Culture ,business - Abstract
The therapy for treatment of Mycobacterium tuberculosis infections is long and arduous. It has been hypothesized that the therapy duration is driven primarily by populations of organisms in different metabolic states that replicate slowly or not at all (acid-phase and nonreplicative-persister [NRP]-phase organisms). Linezolid is an oxazolidinone antimicrobial with substantial activity against Log-phase M. tuberculosis . Here, we examined organisms in acid-phase growth and nonreplicative-persister-phenotype growth and determined the effect of differing clinically relevant exposures to linezolid in a hollow-fiber infection model (HFIM). The endpoints measured were bacterial kill over 29 days and whether organisms that were less susceptible to linezolid could be recovered during that period. In addition, we evaluated the effect of administration schedule on linezolid activity, contrasting daily administration with administration of twice the daily dose every other day. Linezolid demonstrated robust activity when administered daily against both acid-phase and NRP-phase organisms. We demonstrated a clear dose response, with 900 mg of linezolid daily generating ≥3 Log(CFU/ml) killing of acid-phase and NRP-phase M. tuberculosis over 29 days. Amplification of a population less susceptible to linezolid was not seen. Activity was reduced with every 48-h dosing, indicating that the minimum concentration ( C min )/MIC ratio drove the microbiological effect. We conclude that once-daily linezolid dosing has substantial activity against M. tuberculosis in acid-phase and NRP-phase metabolic states. Other studies have shown activity against Log-phase M. tuberculosis . Linezolid is a valuable addition to the therapeutic armamentarium for M. tuberculosis and has the potential for substantially shortening therapy duration.
- Published
- 2018
33. Determination of the Dynamically Linked Indices of Fosfomycin for Pseudomonas aeruginosa in the Hollow Fiber Infection Model
- Author
-
Arnold Louie, Jocelyn Nole, Brandon Duncanson, George L. Drusano, Michael Maynard, and Michael Vicchiarelli
- Subjects
0301 basic medicine ,030106 microbiology ,Microbial Sensitivity Tests ,Fosfomycin ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Anti-Infective Agents ,medicine ,Pharmacology (medical) ,Pseudomonas Infections ,Experimental Therapeutics ,030212 general & internal medicine ,Pharmacology ,biology ,Pseudomonas aeruginosa ,business.industry ,Bacterial pneumonia ,medicine.disease ,Antimicrobial ,biology.organism_classification ,Enterobacteriaceae ,In vitro ,Anti-Bacterial Agents ,Regimen ,Infectious Diseases ,Pharmacodynamics ,business ,medicine.drug - Abstract
Fosfomycin is the only expoxide antimicrobial and is currently under development in the United States as an intravenously administered product. We were interested in identifying the exposure indices most closely linked to its ability to kill bacterial cells and to suppress amplification of less susceptible subpopulations. We employed the hollow fiber infection model for this investigation and studied wild-type strain Pseudomonas aeruginosa PAO1. Because of anticipated rapid resistance emergence, we shortened the study duration to 24 h but sampled the system more intensively. Doses of 12 and 18 g/day and schedules of daily administration, administration every 8 h, and administration by continuous infusion for each daily dose were studied. We measured fosfomycin concentrations (by liquid chromatography-tandem mass spectrometry), the total bacterial burden, and the burden of less susceptible isolates. We applied a mathematical model to all the data simultaneously. There was a rapid emergence of resistance with all doses and schedules. Prior to resistance emergence, an initial kill of 2 to 3 log 10 (CFU/ml) was observed. The model demonstrated that the area under the concentration-time curve/MIC ratio was linked to total bacterial kill, while the time that the concentration remained above the MIC (or, equivalently, the minimum concentration/MIC ratio) was linked to resistance suppression. These findings were also seen in other investigations with Enterobacteriaceae ( in vitro systems) and P. aeruginosa (murine system). We conclude that for serious infections with high bacterial burdens, fosfomycin may be of value as a new therapeutic and may be optimized by administering the agent as a continuous or prolonged infusion or by use of a short dosing interval. For indications such as ventilator-associated bacterial pneumonia, it may be prudent to administer fosfomycin as part of a combination regimen.
- Published
- 2017
34. Combination Treatment With Meropenem Plus Levofloxacin Is Synergistic Against Pseudomonas aeruginosa Infection in a Murine Model of Pneumonia
- Author
-
Alan Schumitzky, Michael Neely, Steven Fikes, David Brown, Michael VanGuilder, Dodge Baluya, Arnold Louie, Roger W. Jelliffe, Stephanie Kurhanewicz, George L. Drusano, Weiguo Liu, and Nichole Robbins
- Subjects
Combination therapy ,Levofloxacin ,Microbial Sensitivity Tests ,Pharmacology ,medicine.disease_cause ,Meropenem ,Microbiology ,Mice ,Major Articles and Brief Reports ,Combined treatment ,Pharmacokinetics ,medicine ,Animals ,Immunology and Allergy ,Pseudomonas Infections ,biology ,business.industry ,Pseudomonas aeruginosa ,Pseudomonas ,Drug Synergism ,Pneumonia ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,In vitro ,Anti-Bacterial Agents ,Disease Models, Animal ,Infectious Diseases ,bacteria ,Drug Therapy, Combination ,Female ,Thienamycins ,business ,medicine.drug - Abstract
Background. Meropenem plus levofloxacin treatment was shown to be a promising combination in our in vitro hollow fiber infection model. We strove to validate this finding in a murine Pseudomonas pneumonia model. Methods. A dose-ranging study with meropenem and levofloxacin alone and in combination against Pseudomonas aeruginosa was performed in a granulocytopenic murine pneumonia model. Meropenem and levofloxacin were administered to partially humanize their pharmacokinetic profiles in mouse serum. Total and resistant bacterial populations were estimated after 24 hours of therapy. Pharmacokinetic profiling of both drugs was performed in plasma and epithelial lining fluid, using a population model. Results. Meropenem and levofloxacin penetrations into epithelial lining fluid were 39.3% and 64.3%, respectively. Both monotherapies demonstrated good exposure responses. An innovative combination-therapy analytic approach demonstrated that the combination was statistically significantly synergistic (α = 2.475), as was shown in the hollow fiber infection model. Bacterial resistant to levofloxacin and meropenem was seen in the control arm. Levofloxacin monotherapy selected for resistance to itself. No resistant subpopulations were observed in any combination therapy arm. Conclusions. The combination of meropenem plus levofloxacin was synergistic, producing good bacterial kill and resistance suppression. Given the track record of safety of each agent, this combination may be worthy of clinical trial.
- Published
- 2014
35. Hollow-Fiber Pharmacodynamic Studies and Mathematical Modeling To Predict the Efficacy of Amoxicillin for Anthrax Postexposure Prophylaxis in Pregnant Women and Children
- Author
-
Weiguo Liu, Arnold Louie, Robert Kulawy, George L. Drusano, and Brian VanScoy
- Subjects
Colony Count, Microbial ,Gene Expression ,Microbial Sensitivity Tests ,Drug resistance ,Pharmacology ,Dicloxacillin ,Drug Administration Schedule ,beta-Lactamases ,Anthrax ,Pregnancy ,Humans ,Medicine ,Computer Simulation ,Drug Dosage Calculations ,Experimental Therapeutics ,Pharmacology (medical) ,Child ,Doxycycline ,Models, Statistical ,biology ,business.industry ,Amoxicillin ,biology.organism_classification ,Anti-Bacterial Agents ,Bacillus anthracis ,Ciprofloxacin ,Regimen ,Infectious Diseases ,Pharmacodynamics ,Female ,business ,Monte Carlo Method ,Half-Life ,medicine.drug - Abstract
Amoxicillin is considered an option for postexposure prophylaxis of Bacillus anthracis in pregnant and postpartum women who are breastfeeding and in children because of the potential toxicities of ciprofloxacin and doxycycline to the fetus and child. The amoxicillin regimen that effectively kills B. anthracis and prevents resistance is unknown. Fourteen-day dose range and dose fractionation studies were conducted in in vitro pharmacodynamic models to identify the exposure intensity and pharmacodynamic index of amoxicillin that are linked with optimized killing of B. anthracis and resistance prevention. Studies with dicloxacillin, a drug resistant to B. anthracis beta-lactamase, evaluated the role of beta-lactamase production in the pharmacodynamic indices for B. anthracis killing and resistance prevention. Dose fractionation studies showed that trough/MIC and not time above MIC was the index for amoxicillin that was linked to successful outcome through resistance prevention. Failure of amoxicillin regimens was due to inducible or stable high level expression of beta-lactamases. Studies with dicloxacillin demonstrated that a time above MIC of ≥94% was linked with treatment success when B. anthracis beta-lactamase activity was negated. Recursive partitioning analysis showed that amoxicillin regimens that produced peak concentrations of 1.75 μg/ml provided a 100% success rate. Other amoxicillin peak and trough values produced success rates of 28 to 67%. For postpartum and pregnant women and children, Monte Carlo simulations predicted success rates for amoxicillin at 1 g every 8 h (q8h) of 53, 33, and 44% (30 mg/kg q8h), respectively. We conclude that amoxicillin is suboptimal for postexposure prophylaxis of B. anthracis in pregnant and postpartum women and in children.
- Published
- 2013
36. Impact of Meropenem in Combination with Tobramycin in a Murine Model of Pseudomonas aeruginosa Pneumonia
- Author
-
Weiguo Liu, David Brown, Arnold Louie, George L. Drusano, and Steven Fikes
- Subjects
Combination therapy ,medicine.medical_treatment ,Population ,Microbial Sensitivity Tests ,Pharmacology ,medicine.disease_cause ,Meropenem ,Mice ,Pharmacokinetics ,Pneumonia, Bacterial ,Tobramycin ,Animals ,Humans ,Medicine ,Drug Interactions ,Experimental Therapeutics ,Pharmacology (medical) ,education ,Lung ,Chemotherapy ,education.field_of_study ,business.industry ,Pseudomonas aeruginosa ,Combination chemotherapy ,Models, Theoretical ,Anti-Bacterial Agents ,Disease Models, Animal ,Treatment Outcome ,Infectious Diseases ,Drug Therapy, Combination ,Female ,Thienamycins ,business ,medicine.drug - Abstract
Pseudomonas aeruginosa pneumonia remains a difficult therapeutic problem. Optimal doses and modes of administration of single agents often do not result in acceptable outcomes. Further, emergence of resistance occurs frequently in this setting with single-agent chemotherapy. The purpose of these experiments was to evaluate combination chemotherapy with meropenem plus tobramycin for P. aeruginosa in a murine pneumonia model. Neutropenia was induced by cyclophosphamide. Pharmacokinetics of meropenem and tobramycin were determined using a population pharmacokinetic approach. Both drugs were given at 4-h intervals. Meropenem was administered as total daily doses of 30 to 600 mg/kg of body weight, while tobramycin doses ranged from 50 to 400 mg/kg. Combination therapy evaluated all combinations of 50, 100, and 150 mg/kg/day of tobramycin doses with 60 or 300 mg/kg/day of meropenem. Total and drug-resistant organisms were enumerated. Meropenem alone had a near-maximal effect at 60 mg/kg/day (3.18 log 10 [CFU/g] kill from stasis). The time > MIC in epithelial lining fluid (ELF) at this dose was 35.25% of 24 h. For tobramycin alone, the near-maximal effect was at 150 mg/kg/day and the area under the concentration-time curve over 24 h in the steady state divided by the MIC (AUC/MIC ratio) in ELF was 240.3. Resistance suppression occurred at an ELF AUC/MIC ratio of 110.6. For combination therapy, the near-maximal effect was reached at 60 mg/kg/day and 50 mg/kg/day of meropenem and tobramycin, which produced a 35.25% time > MIC in ELF and an ELF AUC/MIC ratio of 80.1. The interaction was additive. All combination regimens suppressed resistance. Combination therapy produced additive drug interaction and suppressed all resistance amplification. It is likely that optimal therapy for Pseudomonas aeruginosa pneumonia will involve a combination of agents.
- Published
- 2013
37. Natural History of Yersinia pestis Pneumonia in Aerosol-Challenged BALB/c Mice
- Author
-
Ryan Cirz, Lara Chuvala, George L. Drusano, Renaldo Riggins, Henry S. Heine, Robert T. Cass, Arnold Louie, and Gregory Hurteau
- Subjects
Pneumonic plague ,Yersinia pestis ,Colony Count, Microbial ,Bacteremia ,Spleen ,Bubonic plague ,BALB/c ,Microbiology ,Mice ,Immunity ,medicine ,Animals ,Doubling time ,Experimental Therapeutics ,Pharmacology (medical) ,Lung ,Immune Evasion ,Aerosols ,Pharmacology ,Mice, Inbred BALB C ,Plague ,Models, Statistical ,biology ,biology.organism_classification ,medicine.disease ,Survival Analysis ,Immunity, Innate ,Infectious Diseases ,medicine.anatomical_structure ,Immunology ,Female - Abstract
After a relatively short untreated interval, pneumonic plague has a mortality approaching 100%. We employed a murine model of aerosol challenge with Yersinia pestis to investigate the early course of pneumonic plague in the lung, blood, and spleen. We fit a mathematical model to all data simultaneously. The model fit to the data was acceptable. The number of organisms in the lung at baseline was estimated to be 135 (median) or 1,184 (mean) CFU/g. The doubling time was estimated as 1.5 to 1.7 h. Between 1 and 12 h postexposure, counts declined, but they then increased by 24 h, a finding hypothesized to be due to innate immunity. The model predicted that innate immunity declined with a half-time of 3 to 3.8 h. The threshold for bacteremia was 6.4 × 10 4 to 1.52 × 10 6 CFU/g. By 42 to 48 h, stationary phase was obtained. Lung bacterial burdens exceeded 10 log CFU/g. Obviating early defenses allows for rapid amplification of Y. pestis in bacteremia, making the rapid course with high mortality understandable.
- Published
- 2013
38. Impact of Spores on the Comparative Efficacies of Five Antibiotics for Treatment of Bacillus anthracis in an In Vitro Hollow Fiber Pharmacodynamic Model
- Author
-
David L. Brown, Henry S. Heine, George L. Drusano, Brian VanScoy, Robert Kulawy, and Arnold Louie
- Subjects
medicine.drug_class ,Moxifloxacin ,Antibiotics ,Population ,Microbial Sensitivity Tests ,Models, Biological ,Meropenem ,Microbiology ,chemistry.chemical_compound ,Ciprofloxacin ,Predictive Value of Tests ,Acetamides ,medicine ,Experimental Therapeutics ,Pharmacology (medical) ,education ,Oxazolidinones ,Spores, Bacterial ,Pharmacology ,Doxycycline ,Aza Compounds ,education.field_of_study ,Microbial Viability ,biology ,fungi ,Linezolid ,biochemical phenomena, metabolism, and nutrition ,Viral Load ,bacterial infections and mycoses ,biology.organism_classification ,Virology ,Anti-Bacterial Agents ,Bacillus anthracis ,Infectious Diseases ,chemistry ,Quinolines ,Thienamycins ,Fluoroquinolones ,medicine.drug - Abstract
Bacillus anthracis , the bacterium that causes anthrax, is an agent of bioterrorism. The most effective antimicrobial therapy for B. anthracis infections is unknown. An in vitro pharmacodynamic model of B. anthracis was used to compare the efficacies of simulated clinically prescribed regimens of moxifloxacin, linezolid, and meropenem with the “gold standards,” doxycycline and ciprofloxacin. Treatment outcomes for isogenic spore-forming and non-spore-forming strains of B. anthracis were compared. Against spore-forming B. anthracis , ciprofloxacin, moxifloxacin, linezolid, and meropenem reduced the B. anthracis population by 4 log 10 CFU/ml over 10 days. Doxycycline reduced the population of this B. anthracis strain by 5 log 10 CFU/ml (analysis of variance [ANOVA] P = 0.01 versus other drugs). Against an isogenic non-spore-forming strain, meropenem killed the vegetative B. anthracis the fastest, followed by moxifloxacin and ciprofloxacin and then doxycycline. Linezolid offered the lowest bacterial kill rate. Heat shock studies using the spore-producing B. anthracis strain showed that with moxifloxacin, ciprofloxacin, and meropenem therapies the total population was mostly spores, while the population was primarily vegetative bacteria with linezolid and doxycycline therapies. Spores have a profound impact on the rate and extent of killing of B. anthracis . Against spore-forming B. anthracis , the five antibiotics killed the total (spore and vegetative) bacterial population at similar rates (within 1 log 10 CFU/ml of each other). However, bactericidal antibiotics killed vegetative B. anthracis faster than bacteriostatic drugs. Since only vegetative-phase B. anthracis produces the toxins that may kill the infected host, the rate and mechanism of killing of an antibiotic may determine its overall in vivo efficacy. Further studies are needed to examine this important observation.
- Published
- 2012
39. Evaluation of Once-Daily Vancomycin against Methicillin-Resistant Staphylococcus aureus in a Hollow-Fiber Infection Model
- Author
-
Jürgen B. Bulitta, Thomas P Lodise, Robert Kulawy, Arnold Louie, Anthony M. Nicasio, Rebecca E D'Hondt, and George L. Drusano
- Subjects
Methicillin-Resistant Staphylococcus aureus ,Colony Count, Microbial ,Microbial Sensitivity Tests ,Pharmacology ,Biology ,Staphylococcal infections ,medicine.disease_cause ,Models, Biological ,Microbiology ,Agar plate ,Pharmacokinetics ,Vancomycin ,medicine ,Humans ,Experimental Therapeutics ,Pharmacology (medical) ,Dose fractionation ,Staphylococcal Infections ,medicine.disease ,Methicillin-resistant Staphylococcus aureus ,Anti-Bacterial Agents ,Infectious Diseases ,Staphylococcus aureus ,Area Under Curve ,Pharmacodynamics ,medicine.drug - Abstract
For methicillin-resistant Staphylococcus aureus (MRSA) infections, data suggest that the clinical response is significantly better if the total vancomycin area under the concentration-time curve (AUC)/MIC ratio is ≥400. While the AUC/MIC ratio is the accepted pharmacokinetic/pharmacodynamic (PK/PD) index for vancomycin, this target has been achieved using multiple daily doses. We are unaware of a systematically designed dose fractionation study to compare the bactericidal activity of once-daily administration to that of traditional twice-daily administration. A dose fractionation study was performed with vancomycin in an in vitro hollow-fiber infection model against an MRSA USA300 strain (MIC of 0.75 μg/ml) using an inoculum of ∼10 6 CFU/ml. The three vancomycin regimens evaluated for 168 h were 2 g every 24 h (q24h) as a 1-h infusion, 1 g q12h as a 1-h infusion, and 2 g q24h as a continuous infusion. Free steady-state concentrations (assuming 45% binding) for a total daily AUC/MIC ratio of ≥400 were simulated for all regimens. A validated liquid chromatography-tandem mass spectrometry method was used to determine vancomycin concentrations. Although once-daily and twice-daily dosage regimens exhibited total trough concentrations of
- Published
- 2012
40. Differential Effects of Linezolid and Ciprofloxacin on Toxin Production by Bacillus anthracis in an In Vitro Pharmacodynamic System
- Author
-
David L. Brown, Arnold Louie, Kari Holman, Weiguo Liu, Brian VanScoy, Henry S. Heine, George L. Drusano, Robert Kulawy, and Terry Abshire
- Subjects
Bacterial Toxins ,Population ,Microbial Sensitivity Tests ,medicine.disease_cause ,Models, Biological ,Microbiology ,Anthrax ,chemistry.chemical_compound ,Ciprofloxacin ,Acetamides ,medicine ,heterocyclic compounds ,Experimental Therapeutics ,Pharmacology (medical) ,education ,Infusion Pumps ,Oxazolidinones ,Spores, Bacterial ,Pharmacology ,education.field_of_study ,biology ,Toxin ,organic chemicals ,Linezolid ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,Bacterial Load ,Anti-Bacterial Agents ,Bacillus anthracis ,Spore ,Infectious Diseases ,chemistry ,Pharmacodynamics ,bacteria ,Diffusion Chambers, Culture ,Bacteria ,medicine.drug - Abstract
Bacillus anthracis causes anthrax. Ciprofloxacin is a gold standard for the treatment of anthrax. Previously, using the non-toxin-producing ΔSterne strain of B. anthracis , we demonstrated that linezolid was equivalent to ciprofloxacin for reducing the total (vegetative and spore) bacterial population. With ciprofloxacin therapy, the total population consisted of spores. With linezolid therapy, the population consisted primarily of vegetative bacteria. Linezolid is a protein synthesis inhibitor, while ciprofloxacin is not. Since toxins are produced only by vegetative B. anthracis , the effect of linezolid and ciprofloxacin on toxin production is of interest. The effect of simulated clinical regimens of ciprofloxacin and linezolid on the vegetative and spore populations and on toxin production was examined in an in vitro pharmacodynamic model over 15 days by using the toxin-producing Sterne strain of B. anthracis . Ciprofloxacin and linezolid reduced the total Sterne population at similar rates. With ciprofloxacin therapy, the total Sterne population consisted of spores. With linezolid therapy, >90% of the population was vegetative B. anthracis . With ciprofloxacin therapy, toxin was first detectable at 3 h and remained detectable for at least 5 h. Toxin was never detected with linezolid therapy. Ciprofloxacin and linezolid reduced the total Sterne population at similar rates. However, the B. anthracis population was primarily spores with ciprofloxacin therapy and was primarily vegetative bacteria with linezolid therapy. Toxin production was detected for at least 5 h with ciprofloxacin therapy but was never detected with linezolid treatment. Linezolid may have an advantage over ciprofloxacin for the treatment of B. anthracis infections.
- Published
- 2012
41. Pharmacodynamics of β-Lactamase Inhibition by NXL104 in Combination with Ceftaroline: Examining Organisms with Multiple Types of β-Lactamases
- Author
-
Ian A. Critchley, Brian VanScoy, David Brown, Robert Kulawy, Weiguo Liu, Caroline Grasso, George L. Drusano, Ronald N. Jones, Mariana Castanheira, George A. Williams, Arnold Louie, and Dirk Thye
- Subjects
medicine.drug_class ,Klebsiella pneumoniae ,Cephalosporin ,Microbial Sensitivity Tests ,Drug resistance ,Biology ,Pharmacology ,Models, Biological ,Drug Administration Schedule ,beta-Lactamases ,Microbiology ,Plasmid ,Bacterial Proteins ,Tandem Mass Spectrometry ,Drug Resistance, Bacterial ,Enterobacter cloacae ,medicine ,Humans ,Drug Dosage Calculations ,Pharmacology (medical) ,Beta-Lactamase Inhibitors ,chemistry.chemical_classification ,Enterobacteriaceae Infections ,Drug Synergism ,biology.organism_classification ,Anti-Bacterial Agents ,Cephalosporins ,Klebsiella Infections ,Infectious Diseases ,Enzyme ,chemistry ,Area Under Curve ,Pharmacodynamics ,beta-Lactamase Inhibitors ,Azabicyclo Compounds ,Chromatography, Liquid - Abstract
New broad-spectrum β-lactamases such as KPC enzymes and CTX-M-15 enzymes threaten to markedly reduce the utility of our armamentarium of β-lactam agents, even our most potent drugs, such as carbapenems. NXL104 is a broad-spectrum non-β-lactam β-lactamase inhibitor. In this evaluation, we examined organisms carrying defined β-lactamases and identified doses and schedules of NXL104 in combination with the new cephalosporin ceftaroline, which would maintain good bacterial cell kill and suppress resistance emergence for a clinically relevant period of 10 days in our hollow-fiber infection model. We examined three strains of Klebsiella pneumoniae and one isolate of Enterobacter cloacae. K. pneumoniae 27-908M carried KPC-2, SHV-27, and TEM-1 β-lactamases. Its isogenic mutant, K. pneumoniae 4207J, was “cured” of the plasmid expressing the KPC-2 enzyme. K. pneumoniae 24-1318A carried a CTX-M-15 enzyme, and E. cloacae 2-77C expressed a stably derepressed AmpC chromosomal β-lactamase. Dose-ranging experiments for NXL104 administered as a continuous infusion with ceftaroline at 600 mg every 8 h allowed identification of a 24-h area under the concentration-time curve (AUC) for NXL104 that mediated bactericidal activity and resistance suppression. Dose fractionation experiments identified that “time > threshold” was the pharmacodynamic index linked to cell kill and resistance suppression. Given these results, we conclude that NXL104 combined with ceftaroline on an 8-hourly administration schedule would be optimal for circumstances in which highly resistant pathogens are likely to be encountered. This combination dosing regimen should allow for optimal bacterial cell kill (highest likelihood of successful clinical outcome) and the suppression of resistance emergence.
- Published
- 2012
42. Resistance Emergence Mechanism and Mechanism of Resistance Suppression by Tobramycin for Cefepime for Pseudomonas aeruginosa
- Author
-
Nadzeya Bahniuk, Brian VanScoy, Robert A. Bonomo, Robert Kulawy, Jürgen B. Bulitta, David Brown, George L. Drusano, Sarah M. Drawz, Arnold Louie, Holland DeFiglio, and Steven Fikes
- Subjects
Combination therapy ,medicine.drug_class ,Cefepime ,Blotting, Western ,Cephalosporin ,Microbial Sensitivity Tests ,Drug resistance ,Biology ,medicine.disease_cause ,Models, Biological ,Drug Administration Schedule ,beta-Lactamases ,Microbiology ,Bacterial Proteins ,Drug Resistance, Bacterial ,medicine ,Tobramycin ,Computer Simulation ,Drug Interactions ,Pseudomonas Infections ,Pharmacology (medical) ,RNA, Messenger ,Protein Synthesis Inhibitors ,Pharmacology ,Protein synthesis inhibitor ,Reverse Transcriptase Polymerase Chain Reaction ,Pseudomonas aeruginosa ,Pseudomonas ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,Anti-Bacterial Agents ,Cephalosporins ,Infectious Diseases ,Monte Carlo Method ,medicine.drug - Abstract
The panoply of resistance mechanisms in Pseudomonas aeruginosa makes resistance suppression difficult. Defining optimal regimens is critical. Cefepime is a cephalosporin whose 3′ side chain provides some stability against AmpC β-lactamases. We examined the activity of cefepime against P. aeruginosa wild-type strain PAO1 and its isogenic AmpC stably derepressed mutant in our hollow-fiber infection model. Dose-ranging studies demonstrated complete failure with resistance emergence (both isolates). Inoculum range studies demonstrated ultimate failure for all inocula. Lower inocula failed last (10 days to 2 weeks). Addition of a β-lactamase inhibitor suppressed resistance even with the stably derepressed isolate. Tobramycin combination studies demonstrated resistance suppression in both the wild-type and the stably derepressed isolates. Quantitating the RNA message by quantitative PCR demonstrated that tobramycin decreased the message relative to that in cefepime-alone experiments. Western blotting with AmpC-specific antibody for P. aeruginosa demonstrated decreased expression. We concluded that suppression of β-lactamase expression by tobramycin (a protein synthesis inhibitor) was at least part of the mechanism behind resistance suppression. Monte Carlo simulation demonstrated that a regimen of 2 g of cefepime every 8 h plus 7 mg/kg of body weight of tobramycin daily would provide robust resistance suppression for Pseudomonas isolates with cefepime MIC values up to 8 mg/liter and tobramycin MIC values up to 1 mg/liter. For P. aeruginosa resistance suppression, combination therapy is critical.
- Published
- 2012
43. Preclinical Evaluations To Identify Optimal Linezolid Regimens for Tuberculosis Therapy
- Author
-
Ashley N. Brown, Dodge Baluya, Kalyani Jambunathan, Jaime L. Rodriquez, Jonathan R. Adams, Awewura Kwara, Richard Hafner, George L. Drusano, Arnold Louie, David L. Brown, and Jon C. Mirsalis
- Subjects
Tuberculosis ,Cell Survival ,Antitubercular Agents ,Drug Evaluation, Preclinical ,Drug resistance ,Pharmacology ,Models, Biological ,Microbiology ,Cell Line ,Mycobacterium tuberculosis ,chemistry.chemical_compound ,Pharmacokinetics ,Virology ,Humans ,Medicine ,heterocyclic compounds ,Microbial Viability ,biology ,business.industry ,organic chemicals ,Linezolid ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease ,QR1-502 ,3. Good health ,Clinical trial ,Regimen ,chemistry ,Toxicity ,bacteria ,business ,Research Article - Abstract
Linezolid is an oxazolidinone with potent activity against Mycobacterium tuberculosis. Linezolid toxicity in patients correlates with the dose and duration of therapy. These toxicities are attributable to the inhibition of mitochondrial protein synthesis. Clinically relevant linezolid regimens were simulated in the in vitro hollow-fiber infection model (HFIM) system to identify the linezolid therapies that minimize toxicity, maximize antibacterial activity, and prevent drug resistance. Linezolid inhibited mitochondrial proteins in an exposure-dependent manner, with toxicity being driven by trough concentrations. Once-daily linezolid killed M. tuberculosis in an exposure-dependent manner. Further, 300 mg linezolid given every 12 hours generated more bacterial kill but more toxicity than 600 mg linezolid given once daily. None of the regimens prevented linezolid resistance. These findings show that with linezolid monotherapy, a clear tradeoff exists between antibacterial activity and toxicity. By identifying the pharmacokinetic parameters linked with toxicity and antibacterial activity, these data can provide guidance for clinical trials evaluating linezolid in multidrug antituberculosis regimens., IMPORTANCE The emergence and spread of multidrug-resistant M. tuberculosis are a major threat to global public health. Linezolid is an oxazolidinone that is licensed for human use and has demonstrated potent activity against multidrug-resistant M. tuberculosis. However, long-term use of linezolid has shown to be toxic in patients, often resulting in thrombocytopenia. We examined therapeutic linezolid regimens in an in vitro model to characterize the exposure-toxicity relationship. The antibacterial activity against M. tuberculosis was also assessed for these regimens, including the amplification or suppression of resistant mutant subpopulations by the chosen regimen. Higher exposures of linezolid resulted in greater antibacterial activity, but with more toxicity and, for some regimens, increased resistant mutant subpopulation amplification, illustrating the trade-off between activity and toxicity. These findings can provide valuable insight for designing optimal dosage regimens for linezolid that are part of the long combination courses used to treat multidrug-resistant M. tuberculosis.
- Published
- 2015
44. Suppression of Emergence of Resistance in Pathogenic Bacteria: Keeping Our Powder Dry, Part 2
- Author
-
Arnold Louie, William W. Hope, Alasdair P. MacGowan, and George L. Drusano
- Subjects
0301 basic medicine ,Antibiotic drug ,medicine.drug_class ,030106 microbiology ,Antibiotics ,medicine.disease_cause ,03 medical and health sciences ,Antibiotic resistance ,Drug Resistance, Bacterial ,Enterobacter cloacae ,medicine ,Escherichia coli ,Infection control ,Humans ,Pharmacology (medical) ,Pharmacology ,Resistance (ecology) ,Acinetobacter ,Pathogenic bacteria ,Minireviews ,Bacterial Infections ,Mycobacterium tuberculosis ,Anti-Bacterial Agents ,Klebsiella pneumoniae ,Infectious Diseases ,Risk analysis (engineering) ,Pseudomonas aeruginosa ,Antibiotic Stewardship ,Drug Therapy, Combination ,Business - Abstract
We are in a crisis of bacterial resistance. For economic reasons, most pharmaceutical companies are abandoning antimicrobial discovery efforts, while, in health care itself, infection control and antibiotic stewardship programs have generally failed to prevent the spread of drug-resistant bacteria. At this point, what can be done? The first step has been taken. Governments and international bodies have declared there is a worldwide crisis in antibiotic drug resistance. As discovery efforts begin anew, what more can be done to protect newly developing agents and improve the use of new drugs to suppress resistance emergence? A neglected path has been the use of recent knowledge regarding antibiotic dosing as single agents and in combination to minimize resistance emergence, while also providing sufficient early bacterial kill. In this review, we look at the data for resistance suppression. Approaches include increasing the intensity of therapy to suppress resistant subpopulations; developing concepts of clinical breakpoints to include issues surrounding suppression of resistance; and paying attention to the duration of therapy, which is another important issue for resistance suppression. New understanding of optimizing combination therapy is of interest for difficult-to-treat pathogens like Pseudomonas aeruginosa , Acinetobacter spp., and multidrug-resistant (MDR) Enterobacteriaceae . These lessons need to be applied to our old drugs as well to preserve them and to be put into national and international antibiotic resistance strategies. As importantly, from a regulatory perspective, new chemical entities should have a resistance suppression plan at the time of regulatory review. In this way, we can make the best of our current situation and improve future prospects.
- Published
- 2015
45. Suppression of Emergence of Resistance in Pathogenic Bacteria: Keeping Our Powder Dry, Part 1
- Author
-
Arnold Louie, George L. Drusano, Alasdair P. MacGowan, and William W. Hope
- Subjects
0301 basic medicine ,Antibiotic drug ,Time Factors ,medicine.drug_class ,030106 microbiology ,Antibiotics ,Drug resistance ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Antibiotic resistance ,Enterobacteriaceae ,Drug Resistance, Multiple, Bacterial ,Drug Resistance, Bacterial ,medicine ,Infection control ,Animals ,Humans ,Pharmacology (medical) ,Pharmacology ,Resistance (ecology) ,Bacteria ,Dose-Response Relationship, Drug ,business.industry ,Pathogenic bacteria ,Minireviews ,Bacterial Infections ,Biotechnology ,Anti-Bacterial Agents ,Infectious Diseases ,Risk analysis (engineering) ,Mutation ,Pseudomonas aeruginosa ,Antibiotic Stewardship ,business - Abstract
We are in a crisis of bacterial resistance. For economic reasons, most pharmaceutical companies are abandoning antimicrobial discovery efforts, while, in health care itself, infection control and antibiotic stewardship programs have generally failed to prevent the spread of drug-resistant bacteria. At this point, what can be done? The first step has been taken. Governments and international bodies have declared there is a worldwide crisis in antibiotic drug resistance. As discovery efforts begin anew, what more can be done to protect newly developing agents and improve the use of new drugs to suppress resistance emergence? A neglected path has been the use of recent knowledge regarding antibiotic dosing as single agents and in combination to minimize resistance emergence, while also providing sufficient early bacterial kill. In this review, we look at the data for resistance suppression. Approaches include increasing the intensity of therapy to suppress resistant subpopulations; developing concepts of clinical breakpoints to include issues surrounding suppression of resistance; and paying attention to the duration of therapy, which is another important issue for resistance suppression. New understanding of optimizing combination therapy is of interest for difficult-to-treat pathogens like Pseudomonas aeruginosa , Acinetobacter spp., and multidrug-resistant (MDR) Enterobacteriaceae . These lessons need to be applied to our old drugs to preserve them as well and need to be put into national and international antibiotic resistance strategies. As importantly, from a regulatory perspective, new chemical entities should have a corresponding resistance suppression plan at the time of regulatory review. In this way, we can make the best of our current situation and improve future prospects.
- Published
- 2015
46. Impact of Granulocytes on the Antimicrobial Effect of Tedizolid in a Mouse Thigh Infection Model
- Author
-
Arnold Louie, George L. Drusano, Weiguo Liu, and Robert Kulawy
- Subjects
Drug ,Staphylococcus aureus ,media_common.quotation_subject ,Tetrazoles ,Microbial Sensitivity Tests ,Biology ,Thigh ,medicine.disease_cause ,Staphylococcal infections ,Microbiology ,Mice ,chemistry.chemical_compound ,Anti-Infective Agents ,medicine ,Animals ,Experimental Therapeutics ,Pharmacology (medical) ,Oxazoles ,Oxazolidinones ,media_common ,Pharmacology ,Models, Theoretical ,Staphylococcal Infections ,Prodrug ,medicine.disease ,Organophosphates ,Infectious Diseases ,Cell killing ,medicine.anatomical_structure ,chemistry ,Linezolid ,Female ,Tedizolid ,Granulocytes - Abstract
Tedizolid (TR-700, formerly torezolid) is the active component of the new oxazolidinone prodrug tedizolid phosphate (TR-701). We had previously demonstrated that tedizolid possessed potent antistaphylococcal activity superior to that of linezolid in a neutropenic mouse thigh infection model (A. Louie, W. Liu, R. Kulawy, and G. L. Drusano, Antimicrob. Agents Chemother. 55:3453-3460, 2011). In the current investigation, we used a mouse thigh infection model to delineate the effect of an interaction of TR-700 and granulocytes on staphylococcal cell killing. We compared the antistaphylococcal killing effect of doses of TR-701 equivalent to human exposures ranging from 200 to 3,200 mg/day in both granulocytopenic and normal mice. The mice were evaluated at 24, 48, and 72 h after therapy initiation. In granulocytopenic mice, a clear exposure response in which, depending on the time point of evaluation, stasis was achieved at “human-equivalent” doses of slightly below 2,300 mg/day (at 24 h) to slightly below 2,000 mg/day (at 72 h) was observed. In immune-normal animals, stasis was achieved at human-equivalent doses of slightly greater than 100 mg/day or less. The variance in bacterial cell killing results was attributable to the presence of granulocytes (without drug), the direct effect of TR-700 on Staphylococcus aureus , and the effect of the drug on Staphylococcus aureus mediated through granulocytes. The majority of the bacterial cell killing in normal animals was attributable to the effect of TR-700 mediated through granulocytes. Additional studies need to be undertaken to elucidate the mechanism underlying this observation.
- Published
- 2011
47. In Vivo Pharmacodynamics of Torezolid Phosphate (TR-701), a New Oxazolidinone Antibiotic, against Methicillin-Susceptible and Methicillin-Resistant Staphylococcus aureus Strains in a Mouse Thigh Infection Model
- Author
-
Robert Kulawy, Arnold Louie, George L. Drusano, and Weiguo Liu
- Subjects
Methicillin-Resistant Staphylococcus aureus ,Serum ,Staphylococcus aureus ,medicine.drug_class ,Antibiotics ,Tetrazoles ,Microbial Sensitivity Tests ,Biology ,medicine.disease_cause ,Microbiology ,Methicillin ,Mice ,chemistry.chemical_compound ,Pharmacokinetics ,Acetamides ,medicine ,Animals ,Humans ,Experimental Therapeutics ,Pharmacology (medical) ,Oxazoles ,Oxazolidinones ,Antibacterial agent ,Pharmacology ,Linezolid ,biochemical phenomena, metabolism, and nutrition ,Staphylococcal Infections ,bacterial infections and mycoses ,Methicillin-resistant Staphylococcus aureus ,Organophosphates ,Anti-Bacterial Agents ,Infectious Diseases ,Thigh ,chemistry ,Pharmacodynamics ,Female ,Tedizolid - Abstract
Torezolid phosphate (TR-701) is the phosphate monoester prodrug of the oxazolidinone TR-700 which demonstrates potent in vitro activity against Gram-positive bacteria, including methicillin-susceptible Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA). The pharmacodynamics of TR-701 or TR-700 (TR-701/700) against S. aureus is incompletely defined. Single-dose pharmacokinetic studies were conducted in mice for TR-701/700. Forty-eight-hour dose range and 24-hour dose fractionation studies were conducted in a neutropenic mouse thigh model of S. aureus infection using MRSA ATCC 33591 to identify the dose and schedule of administration of TR-701/700 that was linked with optimized antimicrobial effect. Additional dose range studies compared the efficacies of TR-701/700 and linezolid for one MSSA strain and one community-associated MRSA strain. In dose range studies, TR-701/700 was equally bactericidal against MSSA and MRSA. Mean doses of 37.6 and 66.9 mg/kg of body weight/day of TR-701/700 resulted in stasis and 1 log CFU/g decreases in bacterial densities, respectively, at 24 h, and mean doses of 35.3, 46.6, and 71.1 mg/kg/day resulted in stasis and 1 and 2 log CFU/g reductions, respectively, at 48 h. Linezolid administered at doses as high as 150 mg/kg/day did not achieve stasis at either time point. Dose fractionation studies demonstrated that the area under the concentration-time curve over 24 h in the steady state divided by the MIC (AUC/MIC ratio) was the pharmacodynamic index for TR-701/700 that was linked with efficacy. TR-701/700 was highly active against MSSA and MRSA, in vivo , and was substantially more efficacious than linezolid, although linezolid's top exposure has half the human exposure. Dose fractionation studies showed that AUC/MIC was the pharmacodynamic index linked with efficacy, indicating that once-daily dosing in humans is feasible.
- Published
- 2011
48. Optimization of Aminoglycoside Therapy
- Author
-
Arnold Louie and George L. Drusano
- Subjects
Drug ,medicine.medical_specialty ,media_common.quotation_subject ,Microbial Sensitivity Tests ,Pharmacology ,Kidney ,Drug Administration Schedule ,Nephrotoxicity ,Pharmacokinetics ,Internal medicine ,medicine ,Humans ,Pharmacology (medical) ,Good outcome ,media_common ,business.industry ,Aminoglycoside ,Bayes Theorem ,Liter ,Anti-Bacterial Agents ,Aminoglycosides ,Infectious Diseases ,Mic values ,Toxicity ,business - Abstract
Aminoglycosides are experiencing a resurgence in use because of the spread of multiresistant Gram-negative pathogens. Use of these agents is attended by the occurrence of nephrotoxicity. Aminoglycoside optimization of dose can be defined as the dose having the highest likelihood of a good outcome and the lowest likelihood of toxicity. We have defined the metric Δ as the difference between the likelihoods of good outcome and toxicity, with higher values being better. We developed a method for explicitly evaluating Δ for different daily doses of drug and different schedules of administration. In the empirical therapy setting, when aminoglycosides are administered every 12 h, treatment of infections caused by microbes with MIC values greater than 1 mg/liter cannot attain a high enough likelihood of a good outcome without engendering an unacceptable toxicity likelihood. Daily administration, by decrementing the likelihood of toxicity, allows higher doses to be employed with more acceptable probabilities of toxicity. Obtaining patient-specific information (concentration-time data) allows better identification of the patient's specific pharmacokinetic parameters and dispersion. As these become better identified, optimal doses become rapidly identified so that optimal outcomes are attained. Optimization of therapy for aminoglycosides requires understanding the relationship between exposure and response as well as that between exposure and toxicity. Furthermore, daily administration is much preferred, and stopping therapy as quickly as possible (a week or less may be optimal) will contribute to the ability to optimize therapy.
- Published
- 2011
49. Comparative Efficacies of Candidate Antibiotics against Yersinia pestis in an In Vitro Pharmacodynamic Model
- Author
-
Robert Kulawy, Henry S. Heine, Arnold Louie, David Brown, Weiguo Liu, George L. Drusano, and Brian VanScoy
- Subjects
Yersinia pestis ,medicine.drug_class ,Antibiotics ,Microbial Sensitivity Tests ,Meropenem ,Microbiology ,Moxifloxacin ,Ampicillin ,medicine ,Humans ,Experimental Therapeutics ,Pharmacology (medical) ,Pharmacology ,biology ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Virology ,Anti-Bacterial Agents ,Ciprofloxacin ,Infectious Diseases ,Streptomycin ,Thienamycins ,Gentamicin ,Gentamicins ,medicine.drug - Abstract
Yersinia pestis , the bacterium that causes plague, is a potential agent of bioterrorism. Streptomycin is the “gold standard” for the treatment of plague infections in humans, but the drug is not available in many countries, and resistance to this antibiotic occurs naturally and has been generated in the laboratory. Other antibiotics have been shown to be active against Y. pestis in vitro and in vivo . However, the relative efficacies of clinically prescribed regimens of these antibiotics with streptomycin and with each other for the killing of Yersinia pestis are unknown. The efficacies of simulated pharmacokinetic profiles for human 10-day clinical regimens of ampicillin, meropenem, moxifloxacin, ciprofloxacin, and gentamicin were compared with the gold standard, streptomycin, for killing of Yersinia pestis in an in vitro pharmacodynamic model. Resistance amplification with therapy was also assessed. Streptomycin killed the microbe in one trial but failed due to resistance amplification in the second trial. In two trials, the other antibiotics consistently reduced the bacterial densities within the pharmacodynamic systems from 10 8 CFU/ml to undetectable levels (2 CFU/ml) between 1 and 3 days of treatment. None of the comparator agents selected for resistance. The comparator antibiotics were superior to streptomycin against Y. pestis and deserve further evaluation.
- Published
- 2011
50. Use of an In Vitro Pharmacodynamic Model To Derive a Moxifloxacin Regimen That Optimizes Kill of Yersinia pestis and Prevents Emergence of Resistance
- Author
-
Martina Kinzig-Schippers, Steven Fikes, Arnold Louie, Weiguo Liu, George L. Drusano, Brian VanScoy, David L. Brown, Fritz Sörgel, A. Eichas, K. Files, and Henry S. Heine
- Subjects
Pneumonic plague ,Yersinia pestis ,Moxifloxacin ,Colony Count, Microbial ,Cmax ,Microbial Sensitivity Tests ,Drug resistance ,Biology ,Pharmacology ,Models, Biological ,Bubonic plague ,Drug Administration Schedule ,Microbiology ,Drug Resistance, Bacterial ,medicine ,Humans ,Experimental Therapeutics ,Pharmacology (medical) ,Doxycycline ,Aza Compounds ,Plague ,Dose-Response Relationship, Drug ,biology.organism_classification ,medicine.disease ,Anti-Bacterial Agents ,Treatment Outcome ,Infectious Diseases ,Streptomycin ,Area Under Curve ,Mutation ,Quinolines ,Monte Carlo Method ,Fluoroquinolones ,medicine.drug - Abstract
Yersinia pestis , the causative agent of bubonic, septicemic, and pneumonic plague, is classified as a CDC category A bioterrorism pathogen. Streptomycin and doxycycline are the “gold standards” for the treatment of plague. However, streptomycin is not available in many countries, and Y. pestis isolates resistant to streptomycin and doxycycline occur naturally and have been generated in laboratories. Moxifloxacin is a fluoroquinolone antibiotic that demonstrates potent activity against Y. pestis in in vitro and animal infection models. However, the dose and frequency of administration of moxifloxacin that would be predicted to optimize treatment efficacy in humans while preventing the emergence of resistance are unknown. Therefore, dose range and dose fractionation studies for moxifloxacin were conducted for Y. pestis in an in vitro pharmacodynamic model in which the half-lives of moxifloxacin in human serum were simulated so as to identify the lowest drug exposure and the schedule of administration that are linked with killing of Y. pestis and with the suppression of resistance. In the dose range studies, simulated moxifloxacin regimens of ≥175 mg/day killed drug-susceptible bacteria without resistance amplification. Dose fractionation studies demonstrated that the AUC (area under the concentration-time curve)/MIC ratio predicted kill of drug-susceptible Y. pestis , while the C max (maximum concentration of the drug in serum)/MIC ratio was linked to resistance prevention. Monte Carlo simulations predicted that moxifloxacin at 400 mg/day would successfully treat human infection due to Y. pestis in 99.8% of subjects and would prevent resistance amplification. We conclude that in an in vitro pharmacodynamic model, the clinically prescribed moxifloxacin regimen of 400 mg/day is predicted to be highly effective for the treatment of Y. pestis infections in humans. Studies of moxifloxacin in animal models of plague are warranted.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.