UB Vice President for Research and Economic Development markup
Brian T Tsuji UB Profile
Pharmacy
Professor and Associate Dean, Clinical and Translational Sciences
701 Ellicott S
Buffalo NY 14203
716-881-7543
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Nicholas M. Smith, Justin R. Lenhard, Katie Rose Boissonneault, Cornelia B. Landersdorfer, J??rgen B. Bulitta, Patricia N. Holden, Alan Forrest, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: Clinical Microbiology and Infection, 2020
Authors: Hubert C. Chua, Andy Tse, Nicholas M. Smith, Kari A. Mergenhagen, Raymond Cha, Brian T. Tsuji
Journal-article: International Journal of Antimicrobial Agents, pp. 106269, 2020
Authors: Zarchi E. Sumon, Charles S. Berenson, John A. Sellick, Zackery P. Bulman, Brian T. Tsuji, Kari A. Mergenhagen
Journal-article: Infectious Diseases, vol. 51, pp. 287-292, 2019
Authors: Phillip J. Bergen, Nicholas M. Smith, Tyler B. Bedard, Zackery P. Bulman, Raymond Cha, Brian T. Tsuji
Book-chapter: Advances in Experimental Medicine and Biology, pp. 251-288, 2019
Authors: Zackery P. Bulman, Miao Zhao, Michael J. Satlin, Liang Chen, Barry N. Kreiswirth, Thomas J. Walsh, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: International Journal of Antimicrobial Agents, vol. 52, pp. 114-118, 2018
Authors: Thomas P Lodise, Nicholas M Smith, Patricia Holden, J Nicholas O???Donnell, Tyler Bedard, Robert A Bonomo, Brian T Tsuji
Journal-article: Open Forum Infectious Diseases, vol. 5, pp. S425-S425, 2018
Authors: Iffat Shafiq, Zackery P. Bulman, Sarah L. Spitznogle, Justin E. Osorio, Irene S. Reilly, Alan J. Lesse, Ganapathi I. Parameswaran, Kari A. Mergenhagen, Brian T. Tsuji
Keywords: Daptomycin MRSA bacteremia ceftaroline daptomycin nonsusceptible
Abstract: There is an urgent need to optimize therapeutic options in patients with methicillin-resistant Staphylococcus aureus (MRSA) bacteremia who have failed conventional therapy. Two clinical isolates were obtained from a 68-year-old male with persistent MRSA bacteremia before and after the development of daptomycin nonsusceptibility. The pharmacodynamic activity of monotherapies and combinations of ceftaroline, daptomycin, cefoxitin, nafcillin and vancomycin were evaluated in time-kill experiments versus 108 CFU/mL of the pre- and post-daptomycin nonsusceptible MRSA isolates. Cefoxitin, nafcillin and vancomycin alone or in combination with ceftaroline failed to generate prolonged bactericidal activity against the post-daptomycin nonsusceptible isolate whereas a ceftaroline-daptomycin combination resulted in 6, 24 and 48h log10(CFU/mL) reductions of 3.90, 4.40 and 6.32. Population analysis profiles revealed a daptomycin heteroresistant subpopulation of the pre-daptomycin nonsusceptible MRSA isolate that expanded by >10,000 on daptomycin agar containing 2-16mg/L in the post-daptomycin nonsusceptible isolate. Daptomycin and ceftaroline combinations may be promising against persistent MRSA bacteremia.
PubMedId: 28116950
Journal-article: Infectious Diseases, vol. 49, pp. 410-416, 2017
Authors: Rachel L. Soon, Justin R. Lenhard, Zackery P. Bulman, Patricia N. Holden, Pamela Kelchlin, Judith N. Steenbergen, Lawrence V. Friedrich, Alan Forrest, Brian T. Tsuji
Journal-article: International Journal of Antimicrobial Agents, vol. 49, pp. 25-30, 2017
Authors: Cornelia B Landersdorfer, Jiping Wang, Veronika Wirth, Ke Chen, Keith S Kaye, Brian T Tsuji, Jian Li, Roger L Nation
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 73, pp. 462-468, 2017
Authors: Mohd Hafidz Mahamad Maifiah, Darren J. Creek, Roger L. Nation, Alan Forrest, Brian T. Tsuji, Tony Velkov, Jian Li
Journal-article: Scientific Reports, vol. 7, 2017
Authors: Nicholas M. Smith, Zackery P. Bulman, Arthur O. Sieron, J??rgen B. Bulitta, Patricia N. Holden, Roger L. Nation, Jian Li, Gerard D. Wright, Brian T. Tsuji
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 72, pp. 2297-2303, 2017
Authors: Justin R. Lenhard, Visanu Thamlikitkul, Fernanda P. Silveira, Samira M. Garonzik, Xun Tao, Alan Forrest, Beom Soo Shin, Keith S. Kaye, J??rgen B. Bulitta, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 72, pp. 1415-1420, 2017
Authors: Zackery P. Bulman, Neang S. Ly, Justin R. Lenhard, Patricia N. Holden, J??rgen B. Bulitta, Brian T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 61, 2017
Authors: Justin R. Lenhard, Nicholas M. Smith, Zackery P. Bulman, Xun Tao, Visanu Thamlikitkul, Beom S. Shin, Roger L. Nation, Jian Li, J??rgen B. Bulitta, Brian T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 61, pp. e01268-16, 2017
Authors: Zackery P. Bulman, Ryan C. Dillon, Brian T. Tsuji
Book-chapter: Methods in Pharmacology and Toxicology, pp. 89-108, 2016
Authors: Samira M. Garonzik, Justin R. Lenhard, Alan Forrest, Patricia N. Holden, J??rgen B. Bulitta, Brian T. Tsuji
Journal-article: PLOS ONE, vol. 11, pp. e0156131, 2016
Authors: Rachel L Soon, Justin R Lenhard, Irene Reilly, Tanya Brown, Alan Forrest, Brian T Tsuji
Journal-article: The Journal of Antibiotics, vol. 70, pp. 98-101, 2016
Authors: Rachel L. Soon, Justin R. Lenhard, Zackery P. Bulman, Patricia N. Holden, Pamela Kelchlin, Judith N. Steenbergen, Lawrence V. Friedrich, Alan Forrest, Brian T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 60, pp. 1967-1973, 2016
Authors: Gauri G. Rao, Neang S. Ly, Brian T. Tsuji, J??rgen B. Bulitta, Alan Forrest
Book-chapter: Systems Pharmacology and Pharmacodynamics, pp. 371-402, 2016
Authors: Justin R. Lenhard, J??rgen B. Bulitta, Terry D. Connell, Natalie King-Lyons, Cornelia B. Landersdorfer, Soon-Ee Cheah, Visanu Thamlikitkul, Beom Soo Shin, Gauri Rao, Patricia N. Holden, Thomas J. Walsh, Alan Forrest, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 72, pp. 153-165, 2016
Authors: Thien B Tran, Tony Velkov, Roger L Nation, Alan Forrest, Brian T Tsuji, Phillip J Bergen, Jian Li
Keywords: Nephrotoxicity Pharmacodynamics Pharmacokinetics Polymyxins
Abstract: The polymyxin antibiotics [colistin and polymyxin B (PMB)] are increasingly used as a last-line option for the treatment of infections caused by extensively drug-resistant Gram-negative bacteria. Despite having similar structures and antibacterial activity in vitro, the two clinically available polymyxins have very different pharmacological properties, as colistin (polymyxin E) is intravenously administered to patients in the form of an inactive prodrug colistin methanesulphonate (sodium). This review will discuss recent progress in the pharmacokinetics/pharmacodynamics and toxicity of colistin and PMB, the factors that affect their pharmacological profiles, and the challenges for the effective use of both polymyxins. Strategies are proposed for optimising their clinical utility based upon the recent pharmacological studies in vitro, in animals and patients. In the 'Bad Bugs, No Drugs' era, polymyxins are a critically important component of the antibiotic armamentarium against difficult-to-treat Gram-negative 'superbugs'. Rational approaches to the use of polymyxins must be pursued to increase their effectiveness and to minimise resistance and toxicity.
PubMedId: 27793510
Journal Article: International journal of antimicrobial agents, vol. 48, 2016
Authors: Brian T. Tsuji, Cornelia B. Landersdorfer, Justin R. Lenhard, Soon-Ee Cheah, Visanu Thamlikitkul, Gauri G. Rao, Patricia N. Holden, Alan Forrest, J??rgen B. Bulitta, Roger L. Nation, Jian Li
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 60, pp. 3913-3920, 2016
Authors: Justin R Lenhard, Jonathan S Gall, Jurgen B Bulitta, Visanu Thamlikitkul, Cornelia B Landersdorfer, Alan Forrest, Roger L Nation, Jian Li, Brian T Tsuji
Keywords: Acinetobacter baumannii Carbapenem resistance Polymyxins
Abstract: The objective of this study was to determine the comparative pharmacodynamics of four different carbapenems in combination with polymyxin B (PMB) against carbapenem-resistant Acinetobacter baumannii isolates using time-kill experiments at two different inocula. Two A. baumannii strains (03-149-1 and N16870) with carbapenem minimum inhibitory concentrations (MICs) ranging from 8 to 64mg/L were investigated in 48-h time-kill experiments using starting inocula of 10
PubMedId: 27773498
Journal Article: International journal of antimicrobial agents, vol. 48, 2016
Authors: Justin R Lenhard, Roger L Nation, Brian T Tsuji
Keywords: Acinetobacter baumannii Combinations Klebsiella pneumoniae Polymyxins Pseudomonas aeruginosa Synergy
Abstract: The proliferation of extensively drug-resistant Gram-negative pathogens has necessitated the therapeutic use of colistin and polymyxin B. However, treatment failures with polymyxin monotherapies and the emergence of polymyxin resistance have catalysed the search for polymyxin combinations that synergistically kill polymyxin-susceptible and -resistant organisms. This mini-review examines recent (2011-2016) in vitro and in vivo studies that have attempted to identify synergistic polymyxin combinations against Pseudomonas aeruginosa, Klebsiella pneumoniae and Acinetobacter baumannii. Clinical evidence for the use of combination regimens is also discussed.
PubMedId: 27865626
Journal Article: International journal of antimicrobial agents, vol. 48, 2016
Authors: Vanessa E. Rees, J??rgen B. Bulitta, Antonio Oliver, Brian T. Tsuji, Craig R. Rayner, Roger L. Nation, Cornelia B. Landersdorfer
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 71, pp. 3157-3167, 2016
Authors: Caroline R Brotzki, Kari A Mergenhagen, Zackery P Bulman, Brian T Tsuji, Charles S Berenson
Abstract: Infective endocarditis caused by Proteus mirabilis is a rare and poorly reported disease, with no well-defined effective antibiotic regimen. Here, we present a case of P. mirabilis aortic valve endocarditis. We reviewed prior cases and treatment regimens, and devised effective treatment, which was guided by in vitro sensitivity and synergy testing on the pathogen. Our patient survived without complications or the need for a surgical intervention.
PubMedId: 27797858
Journal Article: BMJ case reports, vol. 2016, 2016
Authors: Gauri G. Rao, Neang S. Ly, John Diep, Alan Forrest, J??rgen B. Bulitta, Patricia N. Holden, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: International Journal of Antimicrobial Agents, vol. 48, pp. 331-336, 2016
Authors: Melinda M Pettigrew, Brian T Tsuji, Janneane F Gent, Yong Kong, Patricia N Holden, Sanjay Sethi, Timothy F Murphy
Abstract: Little is known about the effect of antibiotics on eradication of carriage and development of resistance in Haemophilus influenzae in individuals with chronic obstructive pulmonary disease (COPD). Our goals were to assess antibiotic susceptibilities, prevalence of resistance genes, and development of resistance in H. influenzae and to evaluate the effect of macrolide and fluoroquinolone administration on H. influenzae eradication. Data were from a 15-year longitudinal study of COPD. Genome sequence data were used to determine genotype and identify resistance genes. MICs of antibiotics were determined by reference broth microdilution. Generalized linear mixed models were used to evaluate associations between antibiotic use and H. influenzae eradication. We examined 267 H. influenzae isolates from 77 individuals. All newly acquired H. influenzae isolates were susceptible to azithromycin. Five of 27 (19%) strains developed 4-fold increases in azithromycin MICs and reached or exceeded the susceptibility breakpoint (4 g/ml) during exposure. H. influenzae isolates were uniformly susceptible to ciprofloxacin, levofloxacin, and moxifloxacin (MIC90s of 0.015, 0.015, and 0.06, respectively); there were no mutations in quinolone resistance-determining regions. Fluoroquinolone administration was associated with increased H. influenzae eradication compared to macrolides (odds ratio [OR], 16.67; 95% confidence interval [CI], 2.67 to 104.09). There was no difference in H. influenzae eradication when comparing macrolide administration to no antibiotic (OR, 1.89; 95% CI, 0.43 to 8.30). Fluoroquinolones are effective in eradicating H. influenzae in individuals with COPD. Macrolides are ineffective in eradicating H. influenzae, and their use in COPD patients may lead to decreased macrolide susceptibility and resistance.
PubMedId: 27139476
Journal-article: Antimicrobial agents and chemotherapy, vol. 60, 2016
Authors: Zackery P Bulman, Michael J Satlin, Liang Chen, Barry N Kreiswirth, Beom Soo Shin, Thomas J Walsh, Patricia N Holden, Alan Forrest, Roger L Nation, Jian Li, Brian T Tsuji
Keywords: KPC-producing K. pneumoniae PK/PD meropenem polymyxin B rifampin
Abstract: Pharmacodynamics of a polymyxin B, meropenem, and rifampin triple combination were examined against
PubMedId: 28167549
Journal Article: Antimicrobial agents and chemotherapy, vol. 61, 2016
Authors: Soon-Ee Cheah, Jian Li, Brian T. Tsuji, Alan Forrest, J??rgen B. Bulitta, Roger L. Nation
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 60, pp. 3921-3933, 2016
Authors: Roger L Nation, Jian Li, Otto Cars, William Couet, Michael N Dudley, Keith S Kaye, Johan W Mouton, David L Paterson, Vincent H Tam, Ursula Theuretzbacher, Brian T Tsuji, John D Turnidge
Journal-article: The Lancet Infectious Diseases, vol. 15, pp. 225-234, 2015
Authors: Soon-Ee Cheah, Matthew D Johnson, Yan Zhu, Brian T Tsuji, Alan Forrest, Jurgen B Bulitta, John D Boyce, Roger L Nation, Jian Li
Abstract: Polymyxins are often last-line therapeutic agents used to treat infections caused by multidrug-resistant A. baumannii. Recent reports of polymyxin-resistant A. baumannii highlight the urgent need for research into mechanisms of polymyxin resistance. This study employed genomic and transcriptomic analyses to investigate the mechanisms of polymyxin resistance in A. baumannii AB307-0294 using an in vitro dynamic model to mimic four different clinically relevant dosage regimens of polymyxin B and colistin over 96h. Polymyxin B dosage regimens that achieved peak concentrations above 1mg/L within 1h caused significant bacterial killing (~5 log10CFU/mL), while the gradual accumulation of colistin resulted in no bacterial killing. Polymyxin resistance was observed across all dosage regimens; partial reversion to susceptibility was observed in 6 of 8 bacterial samples during drug-free passaging. Stable polymyxin-resistant samples contained a mutation in pmrB. The transcriptomes of stable and non-stable polymyxin-resistant samples were not substantially different and featured altered expression of genes associated with outer membrane structure and biogenesis. These findings were further supported via integrated analysis of previously published transcriptomics data from strain ATCC19606. Our results provide a foundation for understanding the mechanisms of polymyxin resistance following exposure to polymyxins and the need to explore effective combination therapies.
PubMedId: 27195897
Journal Article: Scientific reports, vol. 6, 2015
Authors: Phillip J Bergen, Zackery P Bulman, Sarith Saju, Juergen B Bulitta, Cornelia Landersdorfer, Alan Forrest, Jian Li, Roger L Nation, Brian T Tsuji
Keywords: colistin colistin methanesulfonate combination polymyxin B polymyxins
Abstract: Since their reintroduction into the clinic in the 1980s, the polymyxin antibiotics colistin-administered intravenously as an inactive prodrug, colistin methanesulfonate (CMS)-and polymyxin B have assumed an important role as salvage therapy for otherwise untreatable gram-negative infections. However, the emerging pharmacodynamic and pharmacokinetic data on CMS/colistin and polymyxin B indicate that polymyxin monotherapy is unlikely to generate plasma concentrations that are reliably efficacious. Additionally, regrowth and the emergence of resistance with monotherapy are commonly reported even when concentrations exceed those achieved clinically. Given this situation, polymyxin combination therapy, which is increasingly being used clinically, has been suggested as a possible means of increasing antimicrobial activity and reducing the development of resistance. Although considerable in vitro data support this view, investigations of polymyxin combination therapy in patients have only recently commenced. The currently available clinical data for polymyxin combinations are generally limited to retrospective analyses and small, low-powered, prospective studies using traditional dosage regimens that achieve low plasma concentrations. Considering the potential for rapid development of resistance to polymyxins, well-designed clinical trials that include higher-dose polymyxin regimens are urgently required to provide a more definitive answer regarding the role of polymyxin combination therapy compared with monotherapy. In this article, we provide an overview of key in vitro and clinical investigations examining CMS/colistin and polymyxin B combination therapy.
PubMedId: 25630411
Journal Article: Pharmacotherapy, vol. 35, 2015
Authors: J??rgen B. Bulitta, Neang S. Ly, Cornelia B. Landersdorfer, Nicholin A. Wanigaratne, Tony Velkov, Rajbharan Yadav, Antonio Oliver, Lisandra Martin, Beom Soo Shin, Alan Forrest, Brian T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 59, pp. 2315-2327, 2015
Authors: Neang S. Ly, J??rgen B. Bulitta, Gauri G. Rao, Cornelia B. Landersdorfer, Patricia N. Holden, Alan Forrest, Phillip J. Bergen, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 70, pp. 1434-1442, 2015
Authors: Phillip J. Bergen, Zackery P. Bulman, Cornelia B. Landersdorfer, Nicholas Smith, Justin R. Lenhard, J??rgen B. Bulitta, Roger L. Nation, Jian Li, Brian T. Tsuji
Journal-article: Infectious Diseases and Therapy, vol. 4, pp. 391-415, 2015
Authors: Zackery P Bulman, Mark D Sutton, Neang S Ly, Jurgen B Bulitta, Patricia N Holden, Roger L Nation, Jian Li, Brian T Tsuji
Abstract: The interplay between polymyxin B pharmacodynamics and pathogenicity was examined in Pseudomonas aeruginosa PAO1 and isogenic DNA repair-deficient mutators (mutM and mutS strains). Against mutS mutators, polymyxin B initial killing was concentration dependent, with >99.9% bacterial reduction at 2 h followed by regrowth and resistance. The pre- versus postexposed strains were inoculated real time into Galleria mellonella waxworms, resulting in increased median survival times from 20 h to 23 h (P < 0.001). Emergence of resistance in mutS P. aeruginosa resulted in attenuation of virulence.
PubMedId: 25918151
Journal-article: Antimicrobial agents and chemotherapy, vol. 59, 2014
Authors: V. E. Rees, J. B. Bulitta, R. L. Nation, B. T. Tsuji, F. Sorgel, C. B. Landersdorfer
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 70, pp. 818-826, 2014
Authors: Justin R Lenhard, Christof von Eiff, Irene S Hong, Patricia N Holden, Michael D Bear, Amy Suen, Zackery P Bulman, Brian T Tsuji
Abstract: Staphylococcus aureus small-colony variants (SCVs) often persist despite antibiotic therapy. Against a 10(8)-CFU/ml methicillin-resistant S. aureus (MRSA) (strain COL) population of which 0%, 1%, 10%, 50%, or 100% was an isogenic hemB knockout (Ia48) subpopulation displaying the SCV phenotype, vancomycin achieved maximal reductions of 4.99, 5.39, 4.50, 3.28, and 1.66 log10 CFU/ml over 48 h. Vancomycin at 16 mg/liter shifted a population from 50% SCV cells at 0 h to 100% SCV cells at 48 h, which was well characterized by a Hill-type model (R2>0.90).
PubMedId: 25451045
Journal Article: Antimicrobial agents and chemotherapy, vol. 59, 2014
Authors: Brian T Tsuji, Yoriko Harigaya, Alan J Lesse, Alan Forrest, Dung Ngo
Abstract: AFN-1252, a potent enoyl-ACP reductase (FabI) inhibitor, is under development for the treatment of Staphylococcus aureus infections. The activity of AFN-1252 against two isolates of S. aureus, MSSA 26213 and MRSA S186, was studied in an in vitro pharmacodynamic model simulating AFN-1252 pharmacokinetics in man. Reductions in bacterial viable count over the first 6 hours were generally 1-2 logs and maximal reductions in viable count were generally achieved at fAUC/MIC ratios of 100-200. Maximum reductions in viable count against MSSA 29213 and MRSA S186 were approximately 4 logs, achieved by 450 mg q12h (fAUC/MIC = 1875) dosing at 28 hours. Staphylococcal resistance to AFN-1252 did not develop throughout the 48-hour experiments. As multidrug resistance continues to increase, these studies support the continued investigation of AFN-1252 as a targeted therapeutic for staphylococcal infections.
PubMedId: 23433442
Journal-article: Journal of Chemotherapy, vol. 25, pp. 32-35, 2013
Authors: Cornelia B. Landersdorfer, Neang S. Ly, Hongmei Xu, Brian T. Tsuji, J??rgen B. Bulitta
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 57, pp. 2343-2351, 2013
Authors: Gauri G Rao, Neang S Ly, Curtis E Haas, Samira Garonzik, Alan Forrest, Jurgen B Bulitta, Pamela A Kelchlin, Patricia N Holden, Roger L Nation, Jian Li, Brian T Tsuji
Abstract: Increasing evidence suggests that colistin monotherapy is suboptimal at currently recommended doses. We hypothesized that front-loading provides an improved dosing strategy for polymyxin antibiotics to maximize killing and minimize total exposure. Here, we utilized an in vitro pharmacodynamic model to examine the impact of front-loaded colistin regimens against a high bacterial density (10(8) CFU/ml) of Pseudomonas aeruginosa. The pharmacokinetics were simulated for patients with hepatic (half-life [t1/2] of 3.2 h) or renal (t1/2 of 14.8 h) disease. Front-loaded regimens (n=5) demonstrated improvement in bacterial killing, with reduced overall free drug areas under the concentration-time curve (fAUC) compared to those with traditional dosing regimens (n=14) with various dosing frequencies (every 12 h [q12h] and q24h). In the renal failure simulations, front-loaded regimens at lower exposures (fAUC of 143 mg h/liter) obtained killing activity similar to that of traditional regimens (fAUC of 268 mg h/liter), with an 97% reduction in the area under the viable count curve over 48 h. In hepatic failure simulations, front-loaded regimens yielded rapid initial killing by up to 7 log10 within 2 h, but considerable regrowth occurred for both front-loaded and traditional regimens. No regimen eradicated the high bacterial inoculum of P. aeruginosa. The current study, which utilizes an in vitro pharmacodynamic infection model, demonstrates the potential benefits of front-loading strategies for polymyxins simulating differential pharmacokinetics in patients with hepatic and renal failure at a range of doses. Our findings may have important clinical implications, as front-loading polymyxins as a part of a combination regimen may be a viable strategy for aggressive treatment of high-bacterial-burden infections.
PubMedId: 24342636
Journal Article: Antimicrobial agents and chemotherapy, vol. 58, 2013
Authors: Hui He, Ji-Chang Li, Roger L Nation, Jovan Jacob, Gong Chen, Hee Ji Lee, Brian T Tsuji, Philip E Thompson, Kade Roberts, Tony Velkov, Jian Li
Keywords: HPLC colistin base activity elemental analysis intravenous administration
Abstract: Very different labelling conventions are employed by different products of colistimethate (CMS), an inactive prodrug of colistin that is used as a last-line defence against Gram-negative 'superbugs'. This study examined the chemical composition and pharmacokinetics in rats of four commercial parenteral products of CMS. Contents per vial of four brands of CMS from three different continents were weighed (n = 3). Elemental analysis and HPLC examination were conducted. The pharmacokinetics of CMS and formed colistin were investigated for each product after intravenous administration in rats (28.1 mg/kg CMS; n = 4). Blood was collected over 180 min, and concentrations of CMS and colistin were measured followed by pharmacokinetic analysis. X-GEN, Paddock and Atlantic products, labelled with 150 mg 'colistin base activity', contained 366.8 0.80, 340.6 0.08 and 380.0 5.97 mg CMS (sodium) per vial, respectively; while the Forest product (labelled with 2 000 000 IU) contained 159.3 1.75 mg CMS (sodium). The elemental compositions of the four products were similar; however, the HPLC profile of the Atlantic CMS was different from those of the other three products. The pharmacokinetics of CMS were generally comparable across brands; however, the molar ratios (%) of the AUC0-180min of colistin to CMS (1.68% 0.35% to 3.29% 0.43%) were significantly different (P = 0.0157). This is the first study to demonstrate that although different brands of CMS from various parts of the world have similar elemental compositions, they lead to different exposures to the microbiologically active formed colistin. The study has significant implications for the interpretation of pharmacological studies of CMS conducted in different parts of the world.
PubMedId: 23749953
Journal Article: The Journal of antimicrobial chemotherapy, vol. 68, 2013
Authors: Brian T Tsuji, Tanya Brown, Ridhi Parasrampuria, Daniel A Brazeau, Alan Forrest, Pamela A Kelchlin, Patricia N Holden, Charles A Peloquin, Debra Hanna, Jurgen B Bulitta
Abstract: Front loading is a strategy used to optimize the pharmacodynamic profile of an antibiotic through the administration of high doses early in therapy for a short duration. Our aims were to evaluate the impact of front loading of linezolid regimens on bacterial killing and suppression of resistance and on RNAIII, the effector molecule of the accessory gene regulator system (encoded by agr) in methicillin-resistant Staphylococcus aureus (MRSA). Time-killing experiments over 48 h were utilized for linezolid against four strains of MRSA: USA100, USA300, USA400, and ATCC 29213. A hollow-fiber infection model simulated traditional and front-loaded human therapeutic regimens of linezolid versus USA300 at 10(6) CFU/ml over 240 h. Over 48 h in time-kill experiments, linezolid displayed bacteriostatic activity, with reductions of >1 log(10) CFU/ml for all strains. Front-loaded regimens that were administered over 5 days, 1,200 mg every 12 h (q12h) (total, 10 doses) and 2,400 mg q12h (total, 10 doses) followed by 300 mg q12h thereafter, resulted in sustained bactericidal activity, with reductions of the area under the CFU curve of -6.15 and -6.03, respectively, reaching undetectable limits at the 10-day study endpoint. All regimens displayed a reduction in RNAIII relative expression at 24 h and 240 h compared with that of the growth control. Monte Carlo simulations predicted a <1.27 increase in the fractional decreases in platelets for all front-loaded regimens versus the 600 mg q12h regimen, except for the highest-dose front-loaded regimen. Front-loading strategies for linezolid are promising and may be of utility in severe MRSA infections, where early aggressive therapy is necessary.
PubMedId: 22526313
Journal Article: Antimicrobial agents and chemotherapy, vol. 56, 2012
Authors: Tohru Takata, Motoyasu Miyazaki, Maki Futo, Shuji Hara, Shouichi Shiotsuka, Hidetoshi Kamimura, Hisae Yoshimura, Akira Matsunaga, Takeshi Nishida, Hiroyasu Ishikura, Takahiko Ishikawa, Kazuo Tamura, Brian T. Tsuji
Journal-article: Scandinavian Journal of Infectious Diseases, vol. 45, pp. 203-212, 2012
Authors: Rachel L Soon, Stuart J Turner, Alan Forrest, Brian T Tsuji, Jack Brown
Abstract: Daptomycin is a novel lipopeptide exhibiting concentration-dependent bactericidal activity against multidrug-resistant Gram-positive pathogens, including MRSA. Approval of daptomycin is granted at 4-6 mg/kg once daily, however off-label use of doses up to 12 mg/kg daily has been utilised without evidence of significant toxicity. Our aim was to optimise daptomycin regimens by assessing the probability of bacteriological efficacy (pTA) and toxicity (pTOX) at various MICs using Monte Carlo simulation (MCS) techniques. Population pharmacokinetic, pharmacodynamic and toxicodynamic models were developed based on current literature. MCS was performed for 10000 patients, who were assigned true weight and creatinine clearances, and were infected with four Staphylococcus aureus strains at each MIC. Bacteriostatic and bactericidal %pTA was calculated following administration of 6, 8, 10 and 12 mg/kg daptomycin; activity was deemed adequate at %pTA 90%. Considerable pharmacodynamic variability was observed in derived AUC/MIC targets between strains. Bacteriostatic targets were adequately attained against all strains at MIC 1 mg/L with daptomycin > 6 mg/kg. However, bactericidal target attainment was only achieved against all strains at the lowest MIC of 0.5 mg/L with daptomycin > 8 mg/kg. At MIC = 2 mg/kg, bactericidal target attainment was extremely poor even at the highest dose of 12 mg/kg. pTOX increased from 3.31% to 17.7% following exposure to 6 mg/kg to 12 mg/kg daily, respectively. Formal benefit:risk analyses favoured doses of 10 mg/kg against infections with MIC < 2 mg/L, whilst modest improvements in activity at 12 mg/kg could not justify the marked increase in pTOX.
PubMedId: 23684388
Journal Article: International journal of antimicrobial agents, vol. 42, 2012
Authors: Zakuan Z Deris, Heidi H Yu, Kathryn Davis, Rachel L Soon, Jovan Jacob, Caron K Ku, Anima Poudyal, Phillip J Bergen, Brian T Tsuji, Jurgen B Bulitta, Alan Forrest, David L Paterson, Tony Velkov, Jian Li, Roger L Nation
Abstract: Multidrug-resistant (MDR) Klebsiella pneumoniae may require combination therapy. We systematically investigated bacterial killing with colistin and doripenem mono- and combination therapy against MDR K. pneumoniae and emergence of colistin resistance. A one-compartment in vitro pharmacokinetic/pharmacodynamic model was employed over a 72-h period with two inocula (10(6) and 10(8) CFU/ml); a colistin-heteroresistant reference strain (ATCC 13883) and three clinical isolates (colistin-susceptible FADDI-KP032 [doripenem resistant], colistin-heteroresistant FADDI-KP033, and colistin-resistant FADDI-KP035) were included. Four combinations utilizing clinically achievable concentrations were investigated. Microbiological responses were examined by determining log changes and population analysis profiles (for emergence of colistin resistance) over 72 h. Against colistin-susceptible and -heteroresistant isolates, combinations of colistin (constant concentration regimens of 0.5 or 2 mg/liter) plus doripenem (steady-state peak concentration [C(max)] of 2.5 or 25 mg/liter over 8 h; half-life, 1.5 h) generally resulted in substantial improvements in bacterial killing at both inocula. Combinations were additive or synergistic against ATCC 13883, FADDI-KP032, and FADDI-KP033 in 9, 9, and 14 of 16 cases (4 combinations at 6, 24, 48, and 72 h) at the 10(6)-CFU/ml inoculum and 14, 11, and 12 of 16 cases at the 10(8)-CFU/ml inoculum, respectively. Combinations at the highest dosage regimens resulted in undetectable bacterial counts at 72 h in 5 of 8 cases (4 isolates at 2 inocula). Emergence of colistin-resistant subpopulations in colistin-susceptible and -heteroresistant isolates was virtually eliminated with combination therapy. Against the colistin-resistant isolate, colistin at 2 mg/liter plus doripenem (C(max), 25 mg/liter) at the low inoculum improved bacterial killing. This investigation provides important information for optimization of colistin-doripenem combinations.
PubMedId: 22802247
Journal Article: Antimicrobial agents and chemotherapy, vol. 56, 2012
Authors: B. T. Tsuji, J. B. Bulitta, T. Brown, A. Forrest, P. A. Kelchlin, P. N. Holden, C. A. Peloquin, L. Skerlos, D. Hanna
Abstract: Vancomycin-resistant enterococci (VRE) have emerged as an important nosocomial pathogen in medical centres worldwide. This study evaluated the impact of front-loading of linezolid on bacterial killing and suppression of resistance against VRE strains with defined genetic mutations. Time-killing experiments over 48 h assessed the concentration effect relationship of linezolid against eight strains of vancomycin-resistant Enterococcus faecalis. A hollow fibre infection model (HFIM) simulated traditional and front-loaded human therapeutic linezolid regimens against VRE strains at 10(6) cfu/mL over 240 h. Translational modelling was performed using S-ADAPT and NONMEM. Over 48 h in time-kill experiments, linezolid displayed bacteriostatic activity with >2 log(10) cfu/mL killing for all strains with an MIC of 4 and minimal activity against VRE with MICs of 16 and 64 mg/L. Against one strain with no resistant alleles (MIC 4 mg/L), 600 mg of linezolid every 12 h achieved maximal reductions of 0.96 log(10) cfu/mL over 240 h in the HFIM, whereas front-loaded 1200 mg of linezolid every 12 h 10 doses or 2400 mg of linezolid every 12 h 10 doses followed by 600 mg of linezolid every 12 h provided significantly improved killing with maximal reductions of 3.02 and 3.46 log(10) cfu/mL. Front-loaded regimens suppressed amplification of resistant subpopulations against VRE strains with no resistant alleles (MIC 4 mg/L) and postponed regrowth of resistant subpopulations against a VRE with 3.2 resistant alleles (MIC 4 mg/L). Modelling yielded excellent population fits (r=0.934) and identified the number of sensitive alleles as a critical covariate. Early, high-dose regimens of linezolid provided promising killing against selected susceptible strains and may be clinically beneficial if early bactericidal activity is necessary.
PubMedId: 22685161
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 67, pp. 2182-2190, 2012
Authors: Neang S Ly, Jenny Yang, Jurgen B Bulitta, Brian T Tsuji
Abstract: The in vitro pharmacodynamics of colistin against Pseudomonas aeruginosa PAO1 wild-type and isogenic knockout strains of phoP and pmrA were evaluated. Colistin killing at subinhibitory concentrations was greater against the phoP and pmrA mutants than the wild type within the first 8 h: the concentration that results in 50% of maximal effect (EC(50)) of the pmrA mutant (0.413 mg/liter) was less than that of the wild type (0.718 mg/liter) (P < 0.05). An in vitro pharmacodynamic model simulating human colistin regimens displayed initial killing followed by regrowth in the phoP mutant and gradual regrowth in the pmrA mutant and wild type.
PubMedId: 22470116
Journal Article: Antimicrobial agents and chemotherapy, vol. 56, 2012
Authors: Phillip J. Bergen, Alan Forrest, J??rgen B. Bulitta, Brian T. Tsuji, Hanna E. Sidjabat, David L. Paterson, Jian Li, Roger L. Nation
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 55, pp. 5134-5142, 2011
Authors: Olanrewaju O. Okusanya, Brian T. Tsuji, Jurgen B. B??litta, Alan Forrest, Catharine C. Bulik, Sujata M. Bhavnani, Prabhavathi Fernandes, Paul G. Ambrose
Journal-article: Diagnostic Microbiology and Infectious Disease, vol. 70, pp. 101-111, 2011
Authors: Yoriko Harigaya, Dung Ngo, Alan J Lesse, Vanthida Huang, Brian T Tsuji
Abstract: The development of hVISA has been associated with vancomycin clinical failures and is commonly misidentified in clinical microbiology laboratories. Therefore, the objectives of this present study was to improve the reliability of methodologies and criteria for identifying hVISA, evaluate the prevalence of hVISA among clinical bloodstream isolates of S. aureus and determine if there exists a relationship between accessory gene regulator (agr) dysfunction and the hVISA phenotype. The presence of hVISA in 220 clinical S. aureus isolates (121 MSSA, 99 MRSA) from bloodstream infections was examined by CLSI broth microdilution, Macro & Standard Etest. Isolates which were classified as hVISA by Macro Etest, were additionally evaluated using a modified PAP-AUC method using a modified starting inoculum of 10(10) CFU/mL, and growth on brain heart infusion agar with 4 mg/L vancomycin (BHIV4) at 10(8) and 10(10) CFU/mL, and agr function was assessed by delta-hemolysin production. Broth microdilution MIC(50/90) of S.aureus and hVISA was 1.0/2.0 and 1.5/2.0 mg/L (p= 0.02), respectively. Macro Etest identified 12 (5.5%) hVISA isolates; higher among MRSA (9.1%) versus MSSA (2.5%) (p = 0.03). The mean modified PAP-AUC ratios (> 0.8) of 7 MRSA strains and 3 MSSA strains were significantly different (p = 0.001). 58% of hVISA strains were found to be agr dysfunctional when 21% of MRSA strains were agr dysfunctional. hVISA was detected among S. aureus bloodstream isolates, which were classified as susceptible among clinical microbiology laboratories. Evaluating the correlation between Etest MICs and modified PAP-AUC ratio values will add further improvement of discriminating hVISA, and agr dysfunction may be predictive of strains which display a greater predilection to display the hVISA phenotype.
PubMedId: 22026752
Journal-article: BMC Infectious Diseases, vol. 11, 2011
Authors: Motoyasu Miyazaki, Tohru Takata, Hisae Yoshimura, Akira Matsunaga, Daiki Ohta, Hiroyasu Ishikura, Maki Futo, Shuji Hara, Hidetoshi Kamimura, Kazuo Tamura, Dung Ngo, Brian T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 55, pp. 1819-1820, 2011
Authors: George Sakoulas, Arnold S. Bayer, Joseph Pogliano, Brian T. Tsuji, Soo-Jin Yang, Nagendra N. Mishra, Victor Nizet, Michael R. Yeaman, Pamela A. Moise
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 56, pp. 838-844, 2011
Authors: Brian T Tsuji, Olanrewaju O Okusanya, Jurgen B Bulitta, Alan Forrest, Sujata M Bhavnani, Prabha B Fernandez, Paul G Ambrose
Abstract: Perhaps the most crucial step in the clinical development of an antimicrobial agent is the selection of a dosing regimen. Such decisions impact not only the success of a program but also the well being of individual patients, the emergence of resistance, and society as a whole. For fusidic acid, the selection of a dosing regimen for the treatment of patients with acute bacterial skin and skin-structure infection (ABSSSI) was based on the integration of knowledge gained from human population pharmacokinetic, in vitro infection, and mathematical models. The overarching goal of these studies was to identify a dosing regimen that would maximize the probabilities of positive clinical outcomes and limit the emergence of bacterial resistance during therapy. Novel dosing regimens identified included 1500 mg twice daily on day 1 followed by 600 mg twice daily for 10-14 days, a regimen that was subsequently found to be effective in a phase 2 clinical study of patients with ABSSSI. Herein, we review the data supporting the use of this novel fusidic acid dosing regimen, which will undergo further clinical evaluation in phase 3 clinical trials.
PubMedId: 21546628
Journal Article: Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, vol. 52 Suppl 7, 2011
Authors: Jill M Butterfield, Brian T Tsuji, Jack Brown, Elizabeth Dodds Ashley, Dwight Hardy, Kristen Brown, Alan Forrest, Thomas P Lodise
Abstract: Despite emerging evidence that dysfunction in the accessory gene regulator (agr) locus is associated with deleterious outcomes among patients treated with vancomycin for methicillin-resistant Staphylococcus aureus (MRSA) infections, factors predictive of agr dysfunction have not been evaluated. This study describes the epidemiology of agr dysfunction, identifies predictors of agr dysfunction in MRSA isolates among those with MRSA bloodstream infections, and describes the relationship between agr dysfunction and other microbiologic phenotypes. A cross-sectional study of patients with MRSA bloodstream infections at two institutions in upstate New York was performed. Clinical data on demographics, comorbidities, disease severity, hospitalization history, and antibiotic history were collected. Microbiologic phenotypes, including agr dysfunction, MIC values by broth microdilution (BMD) and Etest, and vancomycin heteroresistance (hVISA) were tested. Multivariable analyses were performed to identify factors predictive of agr dysfunction. Among 200 patients with an MRSA bloodstream infection, the proportion of strains with agr dysfunction was 31.5%. The distribution of MICs determined by both BMD and Etest were equivalent across agr groups, and there was no association between agr dysfunction and the presence of hVISA. Severity of illness, comorbidities, and hospitalization history were comparable between agr groups. In the multivariate analysis, prior antibiotic exposure was the only factor of variables studied found to be predictive of agr dysfunction. This relationship was predominantly driven by prior beta-lactam and fluoroquinolone administration in the bivariate analysis. Identifying these institution-specific risk factors can be used to develop a process to assess the risk of agr dysfunction and guide empirical antibiotic therapy decisions.
PubMedId: 21930887
Journal Article: Antimicrobial agents and chemotherapy, vol. 55, 2011
Authors: Phillip J Bergen, Brian T Tsuji, Jurgen B Bulitta, Alan Forrest, Jovan Jacob, Hanna E Sidjabat, David L Paterson, Roger L Nation, Jian Li
Abstract: Combination therapy may be required for multidrug-resistant (MDR) Pseudomonas aeruginosa. The aim of this study was to systematically investigate bacterial killing and emergence of colistin resistance with colistin and doripenem combinations against MDR P. aeruginosa. Studies were conducted in a one-compartment in vitro pharmacokinetic/pharmacodynamic model for 96 h at two inocula (~10(6) and ~10(8) CFU/ml) against a colistin-heteroresistant reference strain (ATCC 27853) and a colistin-resistant MDR clinical isolate (19147 n/m). Four combinations utilizing clinically achievable concentrations were investigated. Microbiological response was examined by log changes and population analysis profiles. Colistin (constant concentrations of 0.5 or 2 mg/liter) plus doripenem (peaks of 2.5 or 25 mg/liter every 8 h; half-life, 1.5 h) substantially increased bacterial killing against both strains at the low inoculum, while combinations containing colistin at 2 mg/liter increased activity against ATCC 27853 at the high inoculum; only colistin at 0.5 mg/liter plus doripenem at 2.5 mg/liter failed to improve activity against 19147 n/m at the high inoculum. Combinations were additive or synergistic against ATCC 27853 in 16 and 11 of 20 cases (4 combinations across 5 sample points) at the 10(6)- and 10(8)-CFU/ml inocula, respectively; the corresponding values for 19147 n/m were 16 and 9. Combinations containing doripenem at 25 mg/liter resulted in eradication of 19147 n/m at the low inoculum and substantial reductions in regrowth (including to below the limit of detection at 50 h) at the high inoculum. Emergence of colistin-resistant subpopulations of ATCC 27853 was substantially reduced and delayed with combination therapy. This investigation provides important information for optimization of colistin-doripenem combinations.
PubMedId: 21911563
Journal Article: Antimicrobial agents and chemotherapy, vol. 55, 2011
Authors: Lauren M Lim, Neang Ly, Dana Anderson, Jenny C Yang, Laurie Macander, Anthony Jarkowski, Alan Forrest, Jurgen B Bulitta, Brian T Tsuji
Abstract: Colistin is a polymyxin antibiotic that was discovered in the late 1940s for the treatment of gram-negative infections. After several years of clinical use, its popularity diminished because of reports of significant nephrotoxicity and neurotoxicity. Recently, the antibiotic has resurfaced as a last-line treatment option for multidrug-resistant organisms such as Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae. The need for antibiotics with coverage of these gram-negative pathogens is critical because of their high morbidity and mortality, making colistin a very important treatment option. Unfortunately, however, resistance to colistin has been documented among all three of these organisms in case reports. Although the exact mechanism causing colistin resistance has not been defined, it is hypothesized that the PmrA-PmrB and PhoP-PhoQ genetic regulatory systems may play a role. Colistin dosages must be optimized, as colistin is a last-line treatment option; in addition, suboptimal doses have been linked to the development of resistance. The lack of pharmacokinetic and pharmacodynamic studies and no universal harmonization of dose units, however, have made it difficult to derive optimal dosing regimens and specific dosing guidelines for colistin. In critically ill patients who may have multiorgan failure, renal insufficiency may alter colistin pharmacokinetics. Therefore, dosage alterations in this patient population are imperative to achieve maximal efficacy and minimal toxicity. With regard to colistin toxicity, most studies show that nephrotoxicity is reversible and less frequent than once thought, and neurotoxicity is rare. Further research is needed to fully understand the impact that the two regulatory systems have on resistance, as well as the dosages of colistin needed to inhibit and overcome these developing patterns.
PubMedId: 21114395
Journal Article: Pharmacotherapy, vol. 30, 2010
Authors: Phillip J Bergen, Jurgen B Bulitta, Alan Forrest, Brian T Tsuji, Jian Li, Roger L Nation
Abstract: Colistin plays a key role in treatment of serious infections by Pseudomonas aeruginosa. The aims of this study were to (i) identify the pharmacokinetic/pharmacodynamic (PK/PD) index (i.e., the area under the unbound concentration-time curve to MIC ratio [fAUC/MIC], the unbound maximal concentration to MIC ratio [fC(max)/MIC], or the cumulative percentage of a 24-h period that unbound concentrations exceed the MIC [fT(>MIC)]) that best predicts colistin efficacy and (ii) determine the values for the predictive PK/PD index required to achieve various magnitudes of killing effect. Studies were conducted in a one-compartment in vitro PK/PD model for 24 h using P. aeruginosa ATCC 27853, PAO1, and the multidrug-resistant mucoid clinical isolate 19056 muc. Six intermittent dosing intervals, with a range of fC(max) colistin concentrations, and two continuous infusion regimens were examined. PK/PD indices varied from 0.06 to 18 for targeted fC(max)/MIC, 0.36 to 312 for fAUC/MIC, and 0 to 100% for fT(>MIC). A Hill-type model was fit to killing effect data, which were expressed as the log(10) ratio of the area under the CFU/ml curve for treated regimens versus control. With fC(max) values equal to or above the MIC, rapid killing was observed following the first dose; substantial regrowth occurred by 24 h with most regimens. The overall killing effect was best correlated with fAUC/MIC (R(2) = 0.931) compared to fC(max)/MIC (R(2) = 0.868) and fT(>MIC) (R(2) = 0.785). The magnitudes of fAUC/MIC required for 1- and 2-log(10) reductions in the area under the CFU/ml curve relative to growth control were 22.6 and 30.4, 27.1 and 35.7, and 5.04 and 6.81 for ATCC 27853, PAO1, and 19056 muc, respectively. The PK/PD targets identified will assist in designing optimal dosing strategies for colistin.
PubMedId: 20585118
Journal Article: Antimicrobial agents and chemotherapy, vol. 54, 2010
Authors: J. B. Bulitta, J. C. Yang, L. Yohonn, N. S. Ly, S. V. Brown, R. E. D'Hondt, W. J. Jusko, A. Forrest, B. T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 54, pp. 2051-2062, 2010
Authors: Jurgen B Bulitta, Cornelia B Landersdorfer, Alan Forrest, Silvia V Brown, Michael N Neely, Brian T Tsuji, Arnold Louie
Abstract: Efficacious therapy is of utmost importance to save lives and prevent bacterial resistance in critically ill patients. This review summarizes pharmacokinetic (PK) and pharmacodynamic (PD) modeling methods to optimize clinical care of critically ill patients in empiric and individualized therapy. While these methods apply to all therapeutic areas, we focus on antibiotics to highlight important applications, as emergence of resistance is a significant problem. Nonparametric and parametric population PK modeling, multiple-model dosage design, Monte Carlo simulations, and Bayesian adaptive feedback control are the methods of choice to optimize therapy. Population PK can estimate between patient variability and account for potentially increased clearances and large volumes of distribution in critically ill patients. Once patient- specific PK data become available, target concentration intervention and adaptive feedback control algorithms can most precisely achieve target goals such as clinical cure of an infection or resistance prevention in stable and unstable patients with rapidly changing PK parameters. Many bacterial resistance mechanisms cause PK/PD targets for resistance prevention to be usually several-fold higher than targets for near-maximal killing. In vitro infection models such as the hollow fiber and one-compartment infection models allow one to study antibiotic-induced bacterial killing and emergence of resistance of mono- and combination therapies over clinically relevant treatment durations. Mechanism-based (and empirical) PK/PD modeling can incorporate effects of the immune system and allow one to design innovative dosage regimens and prospective validation studies. Mechanism-based modeling holds great promise to optimize mono- and combination therapy of anti-infectives and drugs from other therapeutic areas for critically ill patients.
PubMedId: 21554212
Journal Article: Current pharmaceutical biotechnology, vol. 12, 2010
Authors: D. Begic, C. von Eiff, B. T. Tsuji
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 63, pp. 977-981, 2009
Authors: Marina Wu, Christof von Eiff, Nahed Al Laham, Brian T Tsuji
Abstract: Catheter-related bloodstream infections due to slow-growing Staphylococcus epidermidis small-colony variants (SCVs) are extremely difficult to treat. Daptomycin and vancomycin pharmacodynamics were evaluated against a site-directed hemB mutant of S. epidermidis displaying the SCV phenotype and compared to that of the parental strain. The maximal killing effect decreased by 7.7-fold for vancomycin and 1.5-fold for daptomycin against the SCV mutant and were well characterized by a Hill-type mathematical model (R2 > 0.97).
PubMedId: 19564372
Journal Article: Antimicrobial agents and chemotherapy, vol. 53, 2009
Authors: Brian T. Tsuji, Yoriko Harigaya, Alan J. Lesse, George Sakoulas, Joseph M. Mylotte
Abstract: The impact of accessory gene regulator (agr) dysfunction and high bacterial density on vancomycin killing and resistance was evaluated among 10 clinical methicillin-resistant Staphylococcus aureus bloodstream isolates using time kill experiments. Under conditions of high inocula and agr dysfunction, vancomycin activity was markedly attenuated, amplifying resistant mutants by 72 h.
PubMedId: 19345040
Journal-article: Diagnostic Microbiology and Infectious Disease, vol. 64, pp. 220-224, 2009
Authors: B. T. Tsuji, J. C. Yang, A. Forrest, P. A. Kelchlin, P. F. Smith
Journal-article: Journal of Antimicrobial Chemotherapy, vol. 62, pp. 156-160, 2008
Authors: Brian T. Tsuji, Steven N. Leonard, Paul R. Rhomberg, Ronald N. Jones, Michael J. Rybak
Journal-article: Diagnostic Microbiology and Infectious Disease, vol. 60, pp. 441-444, 2008
Authors: B. T. Tsuji, C. von Eiff, P. A. Kelchlin, A. Forrest, P. F. Smith
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 52, pp. 1533-1537, 2008
Authors: J. B. Bulitta, N. S. Ly, J. C. Yang, A. Forrest, W. J. Jusko, B. T. Tsuji
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 53, pp. 46-56, 2008
Authors: Masashi Narita, Brian T Tsuji, Victor L Yu
Abstract: Linezolid is an oxazolidinone antibacterial agent indicated for serious gram-positive infections. Only minor adverse effects were seen in phase III trials. However, more serious adverse effects were reported after commercial release, including cases of lactic acidosis, peripheral and optic neuropathy, and serotonin syndrome. Peripheral and optic neuropathy was usually seen after several months of linezolid therapy (median 5 mo), lactic acidosis after several weeks (median 6 wks), and serotonin syndrome after several days (median 4 days). Death occurred in two of seven reported cases of lactic acidosis, and three of 15 reported cases of serotonin syndrome. Improvement or complete recovery occurred in all cases of optic neuropathy, whereas complete recovery failed to occur in any patient with peripheral neuropathy. Linezolid should be discontinued immediately in patients experiencing these adverse effects. Patients receiving linezolid for more than 28 days should be monitored for signs of peripheral and optic neuropathy.
PubMedId: 17655517
Journal Article: Pharmacotherapy, vol. 27, 2007
Authors: Warren E Rose, Michael J Rybak, Brian T Tsuji, Glenn W Kaatz, George Sakoulas
Abstract: Recently, an association between the accessory gene regulator (agr) in Staphylococcus aureus and the development of vancomycin resistance secondary to suboptimal exposure has been demonstrated. We investigated the relationship of vancomycin with and without gentamicin or rifampicin, and daptomycin in the development of resistance in agr groups I and II. S. aureus belonging to agr groups I and II was exposed to varying concentrations of vancomycin and daptomycin simulating an fAUC/MIC of 14-460 and 30-239, respectively, in an in vitro pharmacodynamic model. Vancomycin regimens resulting in fAUC/MIC of 16.1-107.0 resulted in resistance in agr I and agr II knockout strains, whereas regimens resulting in fAUC/MIC of 16.1 resulted in emergence of resistance in agr I- and agr II-positive strains. Overall, agr-null strains demonstrated a higher likelihood of resistance and a greater change in vancomycin susceptibility. The addition of gentamicin and rifampicin to vancomycin at these same exposures prevented the emergence of resistance. At extremely low daptomycin exposures of fAUC/MIC of 22-66, an increase in MIC of 2-3-fold up to a maximum of 0.75 mg/L was observed. However, this was independent of agr group and/or function and still within the susceptible range of daptomycin. The combination of vancomycin with either rifampicin or gentamicin prevented the emergence of vancomycin resistance in agr I and II S. aureus isolates. Changes in daptomycin susceptibility were independent of agr group and function. The association between agr and vancomycin resistance in S. aureus requires further investigation.
PubMedId: 17434881
Journal Article: The Journal of antimicrobial chemotherapy, vol. 59, 2007
Authors: Jenny C Yang, Brian T Tsuji, Alan Forrest
Abstract: Fluoroquinolones (FQs) are broad spectrum, concentration dependent, bactericidal antimicrobials that have been commonly utilized to treat severe nosocomial infections. FQ activity is derived from their ability to inhibit DNA gyrase and topoisomerase IV; resistance has been shown to develop in target site mutations, alterations in efflux pump systems, and incorporation of plasmids. The probability of preventing emergence of resistance and achieving maximal rates of kill are best related to the ratio of free-drug in the area under the concentration-time curve (AUC) to minimum inhibitory concentration (AUC:MIC). Major dosage adjustments for FQs are not necessary in hepatic insufficiency, accumulation of extracellular fluids, and burn patients. Appropriate dosage adjustments in renal function should be taken into consideration. FQ optimization in the critically ill is a multifactorial process that should be individualized to each patient and should take into account the MIC of the pathogen, pharmacokinetic/pharmacodynamic profile of the FQ, and the patient's pathophysiological state.
PubMedId: 18095222
Journal Article: Seminars in respiratory and critical care medicine, vol. 28, 2007
Authors: K. L. LaPlante, M. J. Rybak, B. Tsuji, T. P. Lodise, G. W. Kaatz
Journal-article: Antimicrobial Agents and Chemotherapy, vol. 51, pp. 1315-1320, 2007
Authors: Brian T Tsuji, Michael J Rybak, Kerry L Lau, George Sakoulas
Abstract: Simulated therapeutic vancomycin exposures were evaluated against agr wild-type and knockout Staphylococcus aureus groups I, II, III, and IV using an in vitro pharmacodynamic model. All agr groups developed intermediate resistance to vancomycin after subtherapeutic exposure. The free unbound fraction of the area under the concentration-time curve (fAUC/MIC) required to suppress resistance was fourfold higher (P < 0.001) in agr dysfunctional strains (112 to 169) than that in parent wild-type strains (28).
PubMedId: 17158941
Journal Article: Antimicrobial agents and chemotherapy, vol. 51, 2006
Authors: Brian T Tsuji, Michael J Rybak
Abstract: The ability to maximize bactericidal activity while minimizing toxicity is a therapeutic goal in the treatment of infective endocarditis. We evaluated the impact of administering short-course regimens of gentamicin in combination with daptomycin or vancomycin against one methicillin-susceptible (MSSA 1199) and one methicillin-resistant (MRSA 494) Staphylococcus aureus isolate using an in vitro pharmacodynamic model with simulated endocardial vegetations over 96 h. Human therapeutic dosing regimens for daptomycin (6 and 8 mg/kg of body weight), vancomycin, and gentamicin were simulated. Short-course combination regimens involving gentamicin were administered either as a single 5-mg/kg dose or three 1-mg/kg doses for only the first 24 h and compared to the regimens administered for the full 96-h duration. For all experiments, physiologic conditions of albumin, calcium, and pH were simulated. Both regimens of daptomycin achieved 99.9% kill by 32 h and maintained bactericidal activity against both isolates, which was significantly different from vancomycin, which displayed bacteriostatic activity (P < 0.05). The effects of all short-course regimens of gentamicin were equal to those of the full-duration regimens in combination with daptomycin. Adding three doses of gentamicin (1 mg/kg) to daptomycin resulted in enhancement and bactericidal activity at 24 h against both MRSA and MSSA. The addition of a single dose of gentamicin (5 mg/kg) enhanced or improved the activity of daptomycin and resulted in early bactericidal activity at 4 h against both isolates. The addition of three doses of gentamicin (1 mg/kg) did not improve the activity of vancomycin. However, the addition of a single 5-mg/kg dose of gentamicin to vancomycin resulted in early enhancement at 4 h and 99.9% kill at 32 h for MRSA. These results suggest that a single high dose of gentamicin in combination with daptomycin or vancomycin may be of utility to maximize synergistic and bactericidal activity and minimize toxicity. Further investigation is warranted.
PubMedId: 15980344
Journal Article: Antimicrobial agents and chemotherapy, vol. 49, 2005
Authors: Brian T Tsuji, Michael J Rybak
Abstract: In search for potential synergistic antimicrobial combinations against Staphylococcus aureus isolates, which display heterogeneous resistance to vancomycin, we evaluated the activities of 21 various combinations involving ampicillin/sulbactam, daptomycin, gentamicin, linezolid, quinupristin/dalfopristin, rifampin, and vancomycin by Etest and time-kill methods. A number of combinations demonstrated either synergistic or additive effects against hGISA SA118 and GISA SA179. Agreement between the Etest and time-kill methods for detecting antimicrobial synergy ranged from 66.6% to 71.4%. The Etest method appears promising, and further investigations are warranted.
PubMedId: 16368477
Journal Article: Diagnostic microbiology and infectious disease, vol. 54, 2005
Presented by CUBS © 2024