Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2023 Sep 28;67(10):e00535-23. doi: 10.1128/aac.00535-23

Impact of antibiotic pharmacokinetics in urine on recurrent bacteriuria following treatment of complicated urinary tract infections

David Melnick 1,, Angela K Talley 1, Vipul K Gupta 1, Ian A Critchley 1, Paul B Eckburg 1, Kamal A Hamed 1, Nivedita Bhatt 1, Gary Moore 2, Daren Austin 3, Christopher M Rubino 4, Sujata M Bhavnani 4, Paul G Ambrose 4
Editor: Anne-Catrin Uhlemann5
PMCID: PMC10583661  PMID: 37768311

ABSTRACT

The clinical relevance of bacteriuria following antibiotic treatment of complicated urinary tract infections in clinical trials remains controversial. We evaluated the impact of urine pharmacokinetics on the timing of recurrent bacteriuria in a recently completed trial that compared oral tebipenem pivoxil hydrobromide to intravenous ertapenem. The urinary clearance and urine dwell time of ertapenem were prolonged relative to tebipenem and were associated with a temporal difference in the repopulation of bladder urine with bacteria following treatment, potentially confounding the assessment of efficacy.

KEYWORDS: urinary tract infection, bacteriuria, pharmacokinetics, tebipenem

INTRODUCTION

Discordance between clinical symptom resolution and recurrence of bacteriuria following antibiotic treatment is frequently reported across clinical trials of complicated urinary tract infections, including acute pyelonephritis (cUTI/AP), particularly in patients with underlying genitourinary abnormalities (1 14). Similarly, in clinical practice, recurrent post-treatment bacteriuria in the absence of clinical symptoms (i.e., asymptomatic bacteriuria) is frequent, and current treatment guidelines advocate against additional antibiotic therapy in this setting (15 19). In a systemic review and meta-analysis of recent cUTI/AP clinical trials, lower microbiological response rates were observed in patients treated with carbapenem antibiotics as compared with newer antibiotics, despite similar clinical response rates (20). Recurrence of bacteriuria may be due to the asymptomatic regrowth of persisting pathogens from protected sites in the urinary tract or to recolonization of the bladder urine by exogenous vaginal or urethral flora, as antibiotic concentrations decline to subinhibitory levels after treatment (20 29). To better understand this phenomenon and its clinical relevance, we evaluated the impact of urine pharmacokinetics (PK) on the timing of recurrent bacteriuria and microbiological response in a recently completed Phase 3 clinical trial (ADAPT-PO; ClinicalTrials.gov number, NCT03788967) comparing orally administered tebipenem pivoxil hydrobromide (TBP-PI-HBr 600 mg every 8 hours) to intravenously administered ertapenem (ETP 1,000 mg every 24 hours) in patients with cUTI/AP (3).

TBP-PI-HBr is an orally bioavailable carbapenem that is rapidly converted to tebipenem (TBP), the active moiety, after oral administration (30). Both TBP and ETP demonstrate broad spectrum in vitro activity against gram-positive and gram-negative pathogens, including fluoroquinolone-resistant and extended-spectrum β-lactamase-producing Enterobacterales isolates, with similar distributions of minimum inhibitory concentrations (31 36). However, the antibiotics differ in their PK profiles. TBP demonstrates low protein binding (free drug 58%) with a plasma half-life of 1 hour and a high fraction of the total dose excreted in urine (55%–60%) over 24 hours (37, 38). In contrast, ETP demonstrates high protein binding (free drug 5%) with a plasma half-life of 4–4.5 hours and a lower percentage excreted in urine (41–45%) over 24 hours (39 41). For the ADAPT-PO trial, the treatments were chosen to match plasma PK-pharmacodynamic (PK-PD) measures based on time-dependent microbicidal activity (3, 37 41). The mean duration of antibiotic treatment was similar (8.5–8.7 days). Clinical response rates [mean (%), treatment difference (TBP-ETP); 95% CI] were high and comparable at the end of treatment [EOT, ≥97.9%; 1.4 (−0.1, 3.4)], test of cure [TOC, ≥93.1%; −0.6 (−4.0, 2.8)], and late follow-up assessments [LFU, ≥88.6%; −1.5 (−5.7, 2.6)] (3). In contrast, microbiological response rates declined in both treatment arms after the EOT, with a larger difference between treatment arms at the TOC (median 7 days post-EOT) as compared with the EOT and LFU assessments [EOT, TBP 97.8%, ETP 96.2%; 1.5 (−0.8, 4.1); TOC, TBP 59.5%, ETP 63.5%; −4.5 (−10.8, 1.9); LFU, TBP 57.2%, ETP 58.2%; −1.5 (−7.9, 5.0)] (3). Furthermore, the pathogens recovered from urine at TOC were predominantly of the same genus and species as those recovered from individual patients at study entry and had similar MIC distribution, resistance determinants, and multi-locus sequence type as the baseline pathogens (data on file). These data support the conclusion that the microbiological failures at the TOC assessment were due to the regrowth of persisting pathogens. The larger difference in microbiological response rates at TOC as compared to EOT or LFU assessments was unexpected given the very similar MIC distributions (MIC90 vs Enterobacterales, 0.12 µg/mL) for both antibiotics. We posited that the difference in the timing of recurrent bacteriuria might be related to a difference in the rate of clearance of antibiotics from the urine after the completion of treatment.

The concentration-time profile in urine for TBP following oral administration of a 600 mg dose of TBP-PI-HBr was determined in healthy adult volunteers (38). The concentration-time profile in urine for ETP was modeled based on renal elimination of a 1,000 mg intravenous dose over time, as previously reported (39 41). Due to high protein binding, renal filtration and urinary elimination of ETP are considerably delayed as compared to TBP (Fig. 1). PK simulations demonstrated that urine concentrations of TBP fell below 0.12 µg/mL in virtually all patients within 24–36 hours. In contrast, ETP urine concentrations above 0.12 µg/mL were maintained in approximately 10% of the patients for 5 days and in some patients for more than a week after the last dose.

Fig 1.

Fig 1

Simulated urinary concentrations for ETP, TBP, and imipenem from 30,000 subjects after the last dose of treatment: median, inter-quartile range (IQR, P25/75), inter-decile range (IDR, P10/P90), and inter-ventile range (IVR, P5/P95), with first 1,000 subjects shown.

To further investigate the possible impact of the longer interval required for ETP to drop to subinhibitory urine levels in patients on the occurrence of bacteriuria at TOC assessment, Monte Carlo simulations were performed vs the MIC distribution of the most common Enterobacterales uropathogens (Fig. 2 and 3). The probability of target attainment (PTA) was evaluated by PK-PD population simulation methods. For each treatment regimen, 30,000 virtual subjects were generated. PK parameters were scaled by allometry. Urine concentrations were calculated based on the fraction excreted during the interval of interest (defined as 6 hours) and a constant assumed rate of urine production of 1 mL/kg/h. The fraction of time above MIC was calculated by subject-sampled MIC distribution across all subjects and summarized by drug, pathogen, and MIC.

Fig 2.

Fig 2

PTA in urine vs Escherichia coli, Klebsiella pneumoniae, and Proteus mirabilis in the week following cessation of treatment with TBP-PI-HBr 600 mg q8h and ETP 1,000 mg q24h. A target for PTA of 10% of the week above MIC was used. Pathogen frequency and MIC distributions from references 21, 31, with no correlation assumed.

Fig 3.

Fig 3

PTA in urine for Escherichia coli, Klebsiella pneumoniae, and Proteus mirabilis in the week following cessation of treatment with TBP-PI-HBr 600 mg q8h and ETP 1,000 mg q24h. A target for PTA of 30% of the week above MIC was used. Pathogen frequency and MIC distributions from references 21, 31, with no correlation assumed.

Theoretical PK-PD targets of maintaining a urine concentration of TBP or ETP above a given MIC for 10% or 30% of the 7 days following the cessation of antibiotic therapy were interrogated. These theoretical targets were chosen to provide a quantitative comparison of the proportion of patients with inhibitory urine concentrations of TBP or ETP following the EOT. For a target of 10% urine time above MIC (T > MIC), based on the urine concentration-time profile, ETP demonstrated PTA > 99% at 0.12 µg/mL, the MIC90 for the Enterobacterales pathogens isolated at baseline. In contrast, TBP demonstrated a parallel PTA of ~30% for the 7 days following the EOT. For the theoretical target of maintaining urine concentrations above the MIC90 for 30% of the 7 days following the cessation of antibiotic therapy, ETP PTA was >70% compared with PTA of 0% for TBP.

It is plausible that repopulation of the bladder urine following treatment of a cUTI requires that antibiotic concentrations in urine drop to subinhibitory levels, allowing for persistent pathogens to resume planktonic growth (18, 19, 24, 25). Although urine PD requirements for suppression of bacteriuria have not been defined, the longer dwell time of ETP in urine after the last dose may contribute to the larger difference in microbiologic response seen at the TOC as compared to the EOT or LFU in this clinical trial. These hypothesis-generating data are being tested in an ongoing cUTI clinical trial comparing oral TBP-PI-HBr to intravenously administered imipenem-cilastatin, both with comparable urine PK (Fig. 1).

ACKNOWLEDGMENTS

The authors would like to acknowledge the editorial assistance of Richard Perry, PharmD, in the preparation of this manuscript, which was supported by Spero Therapeutics, Inc., Cambridge, MA.

D.M., A.K.T., V.K.G., I.A.C., P.B.E., K.A.H., and N.B. were employees or consultants of and may have held stock in Spero Therapeutics, Inc. V.K.G. is currently an employee of Arcus Biosciences, Hayward, CA.

G.M. is the owner of Moore Consulting and has received consulting fees from Spero Therapeutics, Inc.

D.A. is an employee of GlaxoSmithKline.

C.M.R., S.M.B., and P.G.A. are employees of Institute for Clinical Pharmacodynamics and have received research support from Spero Therapeutics, Inc.

All authors were involved in the conceptualization and preparation of this manuscript.

Funding for this analysis was provided by Spero Therapeutics, Inc., Cambridge, MA.

Contributor Information

David Melnick, Email: davidmelnickmd@gmail.com.

Anne-Catrin Uhlemann, Columbia University Irving Medical Center, New York, New York, USA .

REFERENCES

  • 1. Mccabe WR, Jackson GG. 1965. Treatment of pyelonephritis: bacterial, drug and host factors in success or failure among 252 patients. N Engl J Med 272:0137–0144. doi: 10.1056/NEJM196505202722002 [DOI] [PubMed] [Google Scholar]
  • 2. Dunne MW, Aronin SI, Das AF, Akinapelli K, Breen J, Zelasky MT, Puttagunta S. 2023. Sulopenem for the treatment of complicated urinary tract infection including pyelonephritis: a phase 3 randomized trial. Clin Infect Dis 76:78–88. doi: 10.1093/cid/ciac704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Eckburg PB, Muir L, Critchley IA, Walpole S, Kwak H, Phelan AM, Moore G, Jain A, Keutzer T, Dane A, Melnick D, Talley AK. 2022. Oral tebipenem pivoxil hydrobromide in complicated urinary tract infection. N Engl J Med 386:1327–1338. doi: 10.1056/NEJMoa2105462 [DOI] [PubMed] [Google Scholar]
  • 4. Kaye KS, Belley A, Barth P, Lahlou O, Knechtle P, Motta P, Velicitat P. 2022. Effect of Cefepime/Enmetazobactam vs piperacillin/tazobactam on clinical cure and microbiological eradication in patients with complicated urinary tract infection or acute pyelonephritis: a randomized clinical trenmetazobactam vs piperacillin/tazoial. JAMA 328:1304–1314. doi: 10.1001/jama.2022.17034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Kaye KS, Bhowmick T, Metallidis S, Bleasdale SC, Sagan OS, Stus V, Vazquez J, Zaitsev V, Bidair M, Chorvat E, Dragoescu PO, Fedosiuk E, Horcajada JP, Murta C, Sarychev Y, Stoev V, Morgan E, Fusaro K, Griffith D, Lomovskaya O, Alexander EL, Loutit J, Dudley MN, Giamarellos-Bourboulis EJ. 2018. Effect of meropenem-vaborbactam vs piperacillin-tazobactam on clinical cure or improvement and microbial eradication in complicated urinary tract infection: the TANGO I randomized clinical trial. JAMA 319:788–799. doi: 10.1001/jama.2018.0438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Kaye KS, Rice LB, Dane AL, Stus V, Sagan O, Fedosiuk E, Das AF, Skarinsky D, Eckburg PB, Ellis-Grosse EJ. 2019. Fosfomycin for injection (ZTI-01) versus piperacillin-tazobactam for the treatment of complicated urinary tract infection including acute pyelonephritis: ZEUS, a phase 2/3 randomized trial. Clin Infect Dis 69:2045–2056. doi: 10.1093/cid/ciz181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Portsmouth S, Bass A, Echols R, Tillotson G. 2021. Heterogeneity of recent phase 3 complicated urinary tract infection clinical trials. Open Forum Infect Dis 8:fab045. doi: 10.1093/ofid/ofab045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Portsmouth S, van Veenhuyzen D, Echols R, Machida M, Ferreira JCA, Ariyasu M, Tenke P, Nagata TD. 2018. Cefiderocol versus imipenem-cilastatin for the treatment of complicated urinary tract infections caused by gram-negative uropathogens: a phase 2, randomised, double-blind, non-inferiority trial. Lancet Infect Dis 18:1319–1328. doi: 10.1016/S1473-3099(18)30554-1 [DOI] [PubMed] [Google Scholar]
  • 9. Wagenlehner FM, Sobel JD, Newell P, Armstrong J, Huang X, Stone GG, Yates K, Gasink LB. 2016. Ceftazidime-avibactam versus doripenem for the treatment of complicated urinary tract infections, including acute pyelonephritis: RECAPTURE, a phase 3 randomized trial program. Clin Infect Dis 63:754–762. doi: 10.1093/cid/ciw378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Wagenlehner FM, Umeh O, Steenbergen J, Yuan G, Darouiche RO. 2015. Ceftolozane-tazobactam compared with levofloxacin in the treatment of complicated urinary-tract infections, including pyelonephritis: a randomised, double-blind, phase 3 trial (ASPECT-cUTI). Lancet 385:1949–1956. doi: 10.1016/S0140-6736(14)62220-0 [DOI] [PubMed] [Google Scholar]
  • 11. Wagenlehner FME, Cloutier DJ, Komirenko AS, Cebrik DS, Krause KM, Keepers TR, Connolly LE, Miller LG, Friedland I, Dwyer JP, EPIC Study Group . 2019. Once-daily plazomicin for complicated urinary tract infections. N Engl J Med 380:729–740. doi: 10.1056/NEJMoa1801467 [DOI] [PubMed] [Google Scholar]
  • 12. Advani SD, Claeys K. 2022. Cefepime/enmetazobactam for complicated urinary tract infections. JAMA 328:1299–1301. doi: 10.1001/jama.2022.15228 [DOI] [PubMed] [Google Scholar]
  • 13. Kadry N, Natarajan M, Bein E, Kim P, Farley J. 2023. Discordant clinical and microbiological outcomes are associated with late clinical relapse in clinical trials for complicated urinary tract infections. Clin Infect Dis 76:1768–1775. doi: 10.1093/cid/ciad010 [DOI] [PubMed] [Google Scholar]
  • 14. McGovern PC, Moeck G, Dane A. 2022. Time to change the complicated urinary tract infection primary Endpoint? the problem and the potential solution. Poster EA01 Presented at the ESCMID/ASM Conference on Drug Development. Dublin, Ireland: [Google Scholar]
  • 15. Cai T, Mazzoli S, Mondaini N, Meacci F, Nesi G, D’Elia C, Malossini G, Boddi V, Bartoletti R. 2012. The role of asymptomatic bacteriuria in young women with recurrent urinary tract infections: to treat or not to treat Clin Infect Dis 55:771–777. doi: 10.1093/cid/cis534 [DOI] [PubMed] [Google Scholar]
  • 16. Nicolle L. 2019. Symptomatic urinary tract infection or asymptomatic bacteriuria? improving care for the elderly. Clin Microbiol Infect 25:779–781. doi: 10.1016/j.cmi.2019.03.013 [DOI] [PubMed] [Google Scholar]
  • 17. Nicolle LE, Gupta K, Bradley SF, Colgan R, DeMuri GP, Drekonja D, Eckert LO, Geerlings SE, Köves B, Hooton TM, Juthani-Mehta M, Knight SL, Saint S, Schaeffer AJ, Trautner B, Wullt B, Siemieniuk R. 2019. Clinical practice guideline for the management of asymptomatic bacteriuria: 2019 update by the infectious diseases society of America. Clin Infect Dis 68:1611–1615. doi: 10.1093/cid/ciz021 [DOI] [PubMed] [Google Scholar]
  • 18. Sobel JD, Brown P. 2020. Urinary tract infections. In Mandell, Douglas, Bennett’s principles and practice of infectious diseases [Google Scholar]
  • 19. Wagenlehner FME, Bjerklund Johansen TE, Cai T, Koves B, Kranz J, Pilatz A, Tandogdu Z. 2020. Epidemiology, definition and treatment of complicated urinary tract infections. Nat Rev Urol 17:586–600. doi: 10.1038/s41585-020-0362-4 [DOI] [PubMed] [Google Scholar]
  • 20. Ezure Y, Rico V, Paterson DL, Hall L, Harris PNA, Soriano A, Roberts JA, Bassetti M, Roberts MJ, Righi E, Wright H. 2022. Efficacy and safety of carbapenems vs new antibiotics for treatment of adult patients with complicated urinary tract infections: a systematic review and meta-analysis. Open Forum Infect Dis 9:faa480. doi: 10.1093/ofid/ofaa480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Stamey TA, Govan DE, Palmer JM. 1965. The localization and treatment of urinary tract infections: the role of bactericidal urine levels as opposed to serum levels. Medicine (Baltimore) 44:1–36. doi: 10.1097/00005792-196501000-00001 [DOI] [PubMed] [Google Scholar]
  • 22. Stamey TA, Fair WR, Timothy MM, Millar MA, Mihara G, Lowery YC. 1974. Serum versus urinary antimicrobial concentrations in cure of urinary-tract infections. N Engl J Med 291:1159–1163. doi: 10.1056/NEJM197411282912204 [DOI] [PubMed] [Google Scholar]
  • 23. Frimodt-Møller N. 2002. Correlation between pharmacokinetic/pharmacodynamic parameters and efficacy for antibiotics in the treatment of urinary tract infection. Int J Antimicrob Agents 19:546–553. doi: 10.1016/s0924-8579(02)00105-x [DOI] [PubMed] [Google Scholar]
  • 24. Chastain DB, King ST, Stover KR. 2018. Rethinking urinary antibiotic breakpoints: analysis of urinary antibiotic concentrations to treat multidrug resistant organisms. BMC Res Notes 11:497. doi: 10.1186/s13104-018-3599-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Conlon BP, Rowe SE, Lewis K. 2015. Persister cells in biofilm associated infections. Adv Exp Med Biol 831:1–9. doi: 10.1007/978-3-319-09782-4_1 [DOI] [PubMed] [Google Scholar]
  • 26. Delcaru C, Alexandru I, Podgoreanu P, Grosu M, Stavropoulos E, Chifiriuc MC, Lazar V. 2016. Microbial biofilms in urinary tract infections and prostatitis: etiology, pathogenicity, and combating strategies. Pathogens 5:65. doi: 10.3390/pathogens5040065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Reid G, Denstedt JD, Kang YS, Lam D, Nause C. 1992. Microbial adhesion and biofilm formation on ureteral stents in vitro and in vivo. J Urol 148:1592–1594. doi: 10.1016/s0022-5347(17)36976-8 [DOI] [PubMed] [Google Scholar]
  • 28. Soto SM. 2014. Importance of biofilms in urinary tract infections: new therapeutic approaches. Adv Biol 2014:1–13. doi: 10.1155/2014/543974 [DOI] [Google Scholar]
  • 29. Tenke P, Köves B, Nagy K, Hultgren SJ, Mendling W, Wullt B, Grabe M, Wagenlehner FME, Cek M, Pickard R, Botto H, Naber KG, Bjerklund Johansen TE. 2012. Update on biofilm infections in the urinary tract. World J Urol 30:51–57. doi: 10.1007/s00345-011-0689-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Jain A, Utley L, Parr TR, Zabawa T, Pucci MJ. 2018. Tebipenem, the first oral carbapenem antibiotic. Expert Rev Anti Infect Ther 16:513–522. doi: 10.1080/14787210.2018.1496821 [DOI] [PubMed] [Google Scholar]
  • 31. Arends SJR, Rhomberg PR, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2019. Antimicrobial activity evaluation of tebipenem (SPR859), an orally available carbapenem, against a global set of enterobacteriaceae isolates, including a challenge set of organisms. Antimicrob Agents Chemother 63:e02618-18. doi: 10.1128/AAC.02618-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Cotroneo N, Rubio A, Critchley IA, Pillar C, Pucci MJ. 2020. In vitro and in vivo characterization of tebipenem, an oral carbapenem. Antimicrob Agents Chemother 64:e02240–19. doi: 10.1128/AAC.02240-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Lacasse E, Brouillette E, Larose A, Parr TR, Rubio A, Malouin F. 2019. in Vitro activity of tebipenem (SPR859) against penicillin-binding proteins of gram-negative and gram-positive bacteria. Antimicrob Agents Chemother 63:e02181-18. doi: 10.1128/AAC.02181-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Mendes RE, Hubler C, Kimbrough JH, Kantro V, Critchley I, Cotroneo N, Castanheira M. 2022. In vitro activity of tebipenem against various resistant subsets of Escherichia coli causing urinary tract infections in the United States. Antimicrob Agents Chemother 66:e0121422. doi: 10.1128/aac.01214-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Arends SJR, Canino MA, Mendes R, Scangarella-Oman NE, Flamm RK. 2020. Comparison of minimum inhibitory concentration results for gepotidacin obtained using agar dilution and broth microdilution methods. Diagn Microbiol Infect Dis 98:115107. doi: 10.1016/j.diagmicrobio.2020.115107 [DOI] [PubMed] [Google Scholar]
  • 36. CLSI . 2018. Performance standards for antimicrobial susceptibility testing; twenty-ninth informational supplement. Vol. M100-S29. Clinical and Laboratory Standards Institute, Wayne, PA. [Google Scholar]
  • 37. McEntee L, Johnson A, Farrington N, Unsworth J, Dane A, Jain A, Cotroneo N, Critchley I, Melnick D, Parr T, Ambrose PG, Das S, Hope W. 2019. Pharmacodynamics of tebipenem: new options for oral treatment of multidrug-resistant gram-negative infections. Antimicrob Agents Chemother 63:e00603-19. doi: 10.1128/AAC.00603-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Eckburg PB, Jain A, Walpole S, Moore G, Utley L, Manyak E, Dane A, Melnick D. 2019. Safety, pharmacokinetics, and food effect of tebipenem pivoxil hydrobromide after single and multiple ascending oral doses in healthy adult subjects. Antimicrob Agents Chemother 63:e00618-19. doi: 10.1128/AAC.00618-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Invanz Prescribing Information, Merck and Company, 2014.
  • 40. Majumdar AK, Musson DG, Birk KL, Kitchen CJ, Holland S, McCrea J, Mistry G, Hesney M, Xi L, Li SX, Haesen R, Blum RA, Lins RL, Greenberg H, Waldman S, Deutsch P, Rogers JD. 2002. Pharmacokinetics of ertapenem in healthy young volunteers. Antimicrob Agents Chemother 46:3506–3511. doi: 10.1128/AAC.46.11.3506-3511.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Chen M, Nafziger AN, Drusano GL, Ma L, Bertino JS. 2006. Comparative pharmacokinetics and pharmacodynamic target attainment of ertapenem in normal-weight, obese, and extremely obese adults. Antimicrob Agents Chemother 50:1222–1227. doi: 10.1128/AAC.50.4.1222-1227.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES