Abstract
The Infectious Diseases Society of America infection-specific guidelines provide limited guidance on the management of focal infections complicated by secondary bacteremias. We address the following 3 commonly encountered questions and management considerations regarding uncomplicated bacteremia not due to Staphylococcus aureus: the role and choice of oral antibiotics focusing on oral beta-lactams, the shortest effective duration of therapy, and the role of repeat blood cultures.
Keywords: bacteremia, oral antibiotics, duration of therapy, repeat blood cultures
Bacteremia complicates approximately 2%–25% of focal infections, such as pneumonia and urinary tract infection (UTI), managed in the hospital and may be associated with poor outcomes [1, 2]. Most infection-specific Infectious Diseases Society of America (IDSA) guidelines do not provide direct management recommendations for choice or route of antibiotic administration, duration of therapy, or use of repeat blood cultures in secondary bacteremias [3, 4]. Until recently, few studies evaluated these common management considerations, and the available literature suggests that considerable practice variation exists [5–7]. Use of the least invasive route of antibiotic administration and the shortest effective duration of therapy are imperative given the relationship between route and duration of antibiotic exposure and adverse drug events, Clostridium difficile infections, and antibiotic resistance [8–11]. Given these considerations, we aim to briefly summarize the existing literature regarding 3 management considerations in uncomplicated bacteremia not due to Staphylococcus aureus in adults: (1) the role and choice of oral antibiotics focusing on beta-lactams, (2) the shortest effective duration of therapy, and (3) the role of repeat blood cultures. No standard definition of uncomplicated bacteremia exists; therefore, we use uncomplicated bacteremia to refer to immunocompetent, bacteremic patients without an uncontrolled source of infection or deep-seated infection for which treatment durations greater than 2 weeks are routinely recommended [3, 12, 13].
WHAT IS THE ROLE OF ORAL ANTIBIOTICS?
Randomized trials, observational studies, and pharmacokinetic-pharmacodynamic principles support the efficacy of high-bioavailability oral agents or agents that achieve approximately equivalent serum concentrations when administered orally or intravenously (eg, fluoroquinolones, oxazolidinones, trimethoprim-sulfamethoxazole) for the treatment of invasive infections including bacteremia from a variety of organisms such as Enterococcus species, Streptococcus spp., Enterobacteriaceae, and Pseudomonas aeruginosa [14–23]. However, antibiotic resistance and the risk of adverse effects often limit the use of these agents, highlighting the need for additional treatment options [24–28]. Oral beta-lactams are well tolerated and retain activity against several relevant organisms that cause bacteremia, such as Enterobacteriaceae and streptococci [25, 28]. However, there are concerns regarding the efficacy of oral beta-lactams for bacteremia because they result in substantially lower serum concentrations compared with intravenous beta-lactams [29, 30]. Despite lower serum concentrations, oral beta-lactams may be effective in specific scenarios given the multifactorial nature and interpatient variability in achieving therapeutic drug concentrations. Unfortunately, there is a lack of robust microbiologic and pharmacokinetic information in varying patient populations to guide the use of oral beta-lactams in many scenarios involving bacteremia.
The IDSA’s community-acquired pneumonia (CAP) guidelines are the only infection-specific guidelines to address the use of oral antibiotics in the setting of bacteremia [4]. The IDSA CAP guidelines suggest that intravenous to oral conversion is safe and effective in the setting of Streptococcus pneumoniae bacteremia [4]. The efficacy of both oral beta-lactams and high-bioavailability oral agents in S. pneumoniae bacteremia from CAP is supported by observational studies and subsets of randomized trials [31–37]. Major areas of uncertainty relate to the use of oral beta-lactams for focal infections with bacteremia caused by Enterobacteriaceae, Enterococcus spp., or Streptococcus spp. apart from pneumonia. Due to an absence of clinical outcomes data regarding use of oral beta-lactams for bacteremia caused by Enterococcus spp. or Streptococcus spp., we will focus on the use of oral beta-lactams for Enterobacteriaceae bacteremia.
There are no randomized controlled trials (RCTs) directly addressing the role of oral beta-lactams in the treatment of Enterobacteriaceae bacteremia; however, some information can be gleaned from RCTs reporting outcomes in the subset of bacteremic patients. First, limited RCT data suggest that use of oral beta-lactams alone without initial intravenous antibiotics is associated with higher clinical and microbiologic recurrences in the setting of complicated UTI and pyelonephritis with or without Enterobacteriaceae bacteremia [38, 39]. Second, clinical cure rates were either consistently greater than 90% or did not differ between bacteremic and nonbacteremic patients in several RCTs involving definitive oral beta-lactam treatment after initial intravenous therapy [40–46]. This observation is based on outcomes directly reported in approximately 50 patients, as well as generic statements in 2 RCTs for which outcomes were not specifically stated [40–46].
Three retrospective cohort studies have more directly investigated the role of oral therapy including beta-lactams in the setting of primarily Enterobacteriaceae bacteremia, with somewhat conflicting results [47–49]. However, reported success rates exceeded 85% in all 3 studies. All 3 studies included a group of patients who received definitive oral therapy after initial intravenous antibiotics for a median of 3–5 days. The most common source of secondary bacteremia was UTI (≥70%), followed by intra-abdominal or biliary infection [47–49]. Mercuro and colleagues performed a single-center study comparing definitive therapy with oral beta-lactams (n = 84) with fluoroquinolones (n = 140) [47]. Clinical success rates were equivalent when comparing oral beta-lactams (86.9%) with fluoroquinolones (87.1%; odds ratio [OR], 1.24; 95% confidence interval [CI], 0.57–2.71) and when intravenous to oral switch occurred within the first 3 days vs later [47]. Kutob and colleagues compared definitive therapy with antibiotics categorized as low (ie, oral beta-lactams, n = 77), moderate (ie, ciprofloxacin, trimethoprim-sulfamethoxazole, n = 179), or high bioavailability (ie, levofloxacin, n = 106) [48]. Failure occurred in 14% of the low, 12% of the moderate, and 2% of the high bioavailability. Both low- (hazard ratio [HR], 7.7; 95% CI, 1.9–51.5) and moderate- (HR, 5.9; 95% CI, 1.6–38.5) compared with high-bioavailability agents were associated with increased risk of failure, and failure occurred earlier in the low bioavailability group [48]. Lastly, Rieger and colleagues performed a study comparing the efficacy of intravenous only (n = 106) vs intravenous to oral treatment (n = 135) for bacteremic UTIs [49]. Treatment failure occurred in 3.8% of the intravenous only vs 8.2% of the intravenous to oral group (P = .19). No specific information was provided regarding outcomes in the 19% of patients receiving oral beta-lactams [49].
There are clinical and pharmacokinetic-pharmacodynamic data supporting the safety and efficacy of high-bioavailability agents (eg, fluoroquinolones, oxazolidinones, trimethoprim-sulfamethoxazole) for the treatment of uncomplicated bacteremia when confirmed by susceptibility testing and in the absence of factors diminishing oral absorption. Additionally, oral beta-lactams can be used for uncomplicated S. pneumoniae bacteremia especially due to pneumonia. Further clinical and pharmacokinetic data are needed to guide the optimal use of oral beta-lactams for uncomplicated Enterobacteriaceae bacteremia; however, available RCT and observational data suggest that conversion from initial intravenous to definitive oral beta-lactam therapy results in high success rates in the appropriately selected patient. It is unclear if high success rates result from the efficacy of oral beta-lactams or the initial course of intravenous antibiotics, as the shortest effective duration of therapy is unknown. Based on the available data, it is reasonable to consider oral beta-lactams as definitive therapy for uncomplicated Enterobacteriaceae bacteremia in patients who have responded clinically to intravenous therapy, particularly in the setting of a pathogen with sufficiently low minimum inhibitory concentration and a patient who is not predisposed to low beta-lactam concentrations (eg, rapid drug elimination, increased volume of distribution). There is a lack of clinical data to guide the use of oral beta-lactams for bacteremia secondary to Streptococcus spp., Enterococcus spp., and alternate infection sources.
WHAT IS THE SHORTEST EFFECTIVE DURATION OF THERAPY?
There is growing evidence that short (≤7 days) as compared with longer treatment durations are equally effective for uncomplicated infections and associated with fewer negative consequences of antibiotic use [8, 10, 50–52]. It is unclear if this evidence applies to patients with secondary bacteremia. The intravascular catheter-related infection guidelines are the IDSA’s only infection-specific guideline to provide a recommendation for duration of therapy in the setting of bacteremia [3]. The recommended duration for uncomplicated Gram-negative bacilli and Enterococcus spp. ranges from 7 to 14 days [3]. Recommendations from non-IDSA guidelines range from 7 to at least 14 days depending on the organism and source [53, 54]. In the absence of clear recommendations, the most commonly used duration is 14 days [5–7, 12].
There are no RCTs published to date comparing durations specifically in bacteremic patients. Limited data exist from RCTs comparing different durations for focal infections and reporting outcomes in the subset of bacteremic patients [55]. A meta-analysis of RCTs comparing the same antibiotic for 5–7 days vs a longer duration identified only 7 trials reporting an outcome in 155 bacteremic patients. The sources of infection were neonatal bacteremia (43%), pneumonia (26%), spontaneous bacterial peritonitis (26%), and pyelonephritis (6%). There were no differences in the rates of clinical cure (risk ratio [RR], 0.88; 95% CI, 0.77–1.01), microbiologic cure (RR, 1.05; 95% CI, 0.91–1.21), or survival (RR, 0.97; 95% CI, 0.76–1.23) [55]. We reviewed additional systematic reviews and meta-analyses of RCTs that included the most common focal infections in hospitalized patients, compared a short (≤7 days) and long (>7 days) duration of therapy, and reported outcomes in the subset of bacteremic patients [55–62]. Six complicated UTI or pyelonephritis RCTs included approximately 140 bacteremic patients [16, 46, 63–66]. A fluoroquinolone was used for a short duration in 5 RCTs, and intravenous to oral beta-lactam was used in 1 RCT. There were no reported differences in clinical cure rates between patients treated for a short or long duration or between bacteremic and nonbacteremic patients [16, 46, 57, 63–66]. Six nonazithromycin CAP RCTs included approximately 90 patients with S. pneumoniae bacteremia, with no differences in clinical efficacy in patients treated with a short vs long duration or in bacteremic vs nonbacteremic patients [31, 35, 67–70]. A short course treatment consisted of a beta-lactam in 3 trials, fluoroquinolone in 2 trials, and ketolide in 1 trial [31, 35, 67–70]. Consistent with the previously cited meta-analysis, there are few bacteremic patients with outcomes available to compare short vs longer duration; however, available RCTs suggest that a shorter duration is as effective as longer durations in Enterobacteriaceae bacteremia secondary to UTIs and S. pneumoniae bacteremia secondary to CAP.
Four published retrospective studies compared a short vs long duration for secondary bacteremias [12, 13, 71, 72]. Chotiprasitsakul and colleagues performed a multicenter, propensity score–matched cohort study comparing 30-day mortality in patients with Enterobacteriaceae bacteremia who received antibiotics for 6 to 10 days (n = 385) vs 11 to 16 days (n = 385) [71]. Median durations were (interquartile range [IQR]) 8 (7–9) and 15 (13–15) days. UTI was the most common source (37%). Short course was not associated with increased 30-day mortality (9.6% vs 10.1%; HR, 1.00; 95% CI, 0.62–1.63) or 30-day recurrent bloodstream infections (1.3% vs 2.3%; OR, 1.32; 95% CI, 0.48–3.41) [71]. Nelson and colleagues performed a multicenter cohort study comparing clinical failure in patients with Gram-negative bacteremia receiving antibiotics for 7–10 days (n = 117) vs longer (n = 294) [13]. Median durations (IQR) were 9 (7–10) and 13 (12–15) days. Urine was the most common source (69%), and Enterobacteriaceae was the most common pathogen (90%). Treatment failure was associated with shorter course therapy (HR, 2.60; 95% CI, 1.20–5.53), with the difference driven by 90-day mortality (8.2% vs 3.3%, P = .04), not 90-day recurrent infection (6.7% vs 6.5%, P = .93). Median time to treatment failure (IQR) was 36 (10–69) days [13]. Daneman and colleagues performed a multicenter, propensity score–matched cohort study in critically ill patients with uncomplicated bacteremia from a wide distribution of sources and pathogens. Two hundred twenty-two matched pairs were included. The median durations (IQR) were 7 (4–8) and 15 days (14–20). Mortality (27% vs 29%; RR, 0.94; 95% CI, 0.70–1.26) and recurrent bacteremia (6% vs 8%, P = .29) were not different in patients receiving short and long durations, respectively [12]. Lastly, Doi and colleagues performed a retrospective single-center study comparing 30-day mortality in patients with bacteremia secondary to cholangitis who received antibiotics for 7 or fewer days (n = 86) compared with longer (n = 177) [72]. All patients had source control, and median durations of therapy were 6 and 12 days. The most common organisms were Gram-negatives (87% vs 89%), but 13% and 27% had polymicrobial bacteremia in the short and long groups, respectively. The 30-day mortality rates were 5% and 6% (OR, 0.82; 95% CI, 0.18–2.95) [72]. In summary, the 3 studies assessing mortality within 30 days or less reported no difference between the different treatment durations [12, 71, 72], while the lone study assessing outcomes within 90 days did identify a difference [13]. While the relative merits of each outcome time frame have been debated, using a 90-day end point increases the likelihood of capturing mortality related to underlying comorbidities rather than the duration of antibiotic treatment for an acute bacteremia [73, 74]. Additional more limited evidence suggests that shorter durations may be as effective as longer durations for a variety of bacteremic sources [47, 75–80].
In summary, the optimal duration of therapy for uncomplicated bacteremia is understudied. More data are needed as a basis for the shortest effective duration. There are multiple ongoing RCTs comparing 7 vs 14 days in patients with bacteremia from various sources and organisms [81–85]. Until results are available, available clinical trial and observational literature suggest that shorter treatment durations are as safe and effective as longer durations for uncomplicated Enterobacteriaceae bacteremia and S. pneumoniae bacteremia from pneumonia. There is a lack of comparative data investigating the optimal treatment duration for non-Enterobacteriaceae Gram-negative organisms (eg, P. aeruginosa, Acinetobacter spp.), Enterococcus spp., and Streptococcus spp., as these organisms were not present or were present in relatively lower numbers in the previously discussed studies. Of note, there are limited published data to support the common practice of treating for 14 days for most clinical scenarios of uncomplicated bacteremia not due to S. aureus.
WHAT IS THE ROLE OF REPEAT BLOOD CULTURES?
With the exception of S. aureus bacteremia, the utility of repeat blood cultures is not well defined. Studies examining this question are small, single-center retrospective studies [86–88]. However, the small amount of existing data suggest limited utility in repeat blood cultures in cases of Gram-negative bacteremia or bacteremia secondary to UTIs and skin and soft tissue infections (SSTIs) [86–88]. The largest study to date investigating the utility of repeat cultures in bacteremia included 701 repeat blood cultures, with persistent bacteremia reported in 17% [86]. Persistent bacteremia was defined as repeat blood culture positivity with the same organism 2–7 days following the initial culture. Of the persistent bacteremias, 76% were Gram-positive organisms, and 54% were from endovascular or bone and joint sources. A nested case-control study was performed comparing patients with persistent (n = 118) vs cleared bacteremia (n = 118). Gram-positive organism, endovascular source, and epidural source were associated with persistent bacteremia in multivariate analysis. Genitourinary source, Escherichia coli, and streptococci were associated with a lower risk [86]. An additional retrospective study performed by Canzoneri and colleagues included 383 repeat blood cultures with an overall positive yield of 14% [87]. Seventy-eight percent of repeat positives were Gram-positive cocci, while 15% were Gram-negatives alone. There was a negative association with persistent bacteremia and UTI or SSTI source. Five follow-up blood cultures were needed to detect 1 positive, but this number increased to 17 when looking only at the Gram-negative cases [87]. A smaller study with 38 repeat blood cultures from bacteremic UTIs showed a repeat positive yield of only 8%, and all repeat positives were secondary to Gram-positive organisms [88]. Of note, prescribed antibiotic durations were significantly longer in patients with repeat blood cultures performed (15 vs 12 days) [88]. The necessity of repeat blood cultures is further called in to question when examining evidence showing the limited clinical utility of initial blood cultures in the cases of S. pneumoniae pneumonia and pyelonephritis [89–91]. In these retrospective studies, bacteremia was not correlated with increased mortality or morbidity [89–91]. Additionally, the results of initial positive blood cultures had no effect on treatment choice in the case of UTI [91].
Given the low yield of repeat blood cultures in uncomplicated non–S. aureus bacteremia, the unclear impact on clinical decision-making, and the potential correlation with increased antibiotic days, there appears to be limited added value in the clinical practice of routinely obtaining repeat blood cultures for the purpose of documenting bloodstream clearance. Therefore, we suggest considering the source of infection when deciding whether to repeat blood cultures because the available literature suggests that repeat blood cultures are low yield in the setting of bacteremia secondary to UTIs and SSTIs. Similarly, we would discourage routine documentation of blood culture clearance with Gram-negative bacteremia. We suggest obtaining repeat blood cultures when the source of bacteremia is unknown or there is a lack of clinical improvement, raising concern for complicated infection. Further studies focusing on specific organisms and sources of infectious would be beneficial given the low number of organisms such as P. aeruginosa in these studies.
SUMMARY
There is currently a lack of extensive evidence to establish strong recommendations for common management considerations in uncomplicated bacteremia. However, it is imperative that new management approaches be considered that balance optimizing clinical outcomes and limiting unintended consequences of excessive antibiotic use. To that end, the available evidence suggests that short treatment durations are safe and effective in the setting of uncomplicated Enterobacteriaceae and S. pneumoniae bacteremia, and there appears to be limited utility for routine repeat blood cultures to document bloodstream clearance in the setting of clinical improvement and minimal concern for complicated infection. High-bioavailability oral agents can be reliably used for uncomplicated bacteremia, and oral beta-lactams can be considered after initial intravenous treatment for select patients with uncomplicated Enterobacteriaceae or S. pneumoniae bacteremia.
Acknowledgments
Financial support. This work was not supported by any funding.
Potential conflicts of interest. All authors: no reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
References
- 1. Coburn B, Morris AM, Tomlinson G, Detsky AS. Does this adult patient with suspected bacteremia require blood cultures?JAMA 2012; 308:502–11. [DOI] [PubMed] [Google Scholar]
- 2. Søgaard M, Nørgaard M, Pedersen L, et al. . Blood culture status and mortality among patients with suspected community-acquired bacteremia: a population-based cohort study. BMC Infect Dis 2011; 11:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Mermel LA, Allon M, Bouza E, et al. . Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infection: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis 2009; 49:1–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Mandell LA, Wunderink RG, Anzueto A, et al. ; Infectious Diseases Society of America; American Thoracic Society Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clin Infect Dis 2007; 44(Suppl 2):S27–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Corona A, Bertolini G, Ricotta AM, et al. . Variability of treatment duration for bacteraemia in the critically ill: a multinational survey. J Antimicrob Chemother 2003; 52:849–52. [DOI] [PubMed] [Google Scholar]
- 6. Daneman N, Shore K, Pinto R, Fowler R. Antibiotic treatment duration for bloodstream infections in critically ill patients: a national survey of Canadian infectious diseases and critical care specialists. Int J Antimicrob Agents 2011; 38:480–5. [DOI] [PubMed] [Google Scholar]
- 7. Diallo K, Thilly N, Luc A, et al. . Management of bloodstream infections by infection specialists: an international ESCMID cross-sectional survey. Int J Antimicrob Agents. 2018; 51:794–8. [DOI] [PubMed] [Google Scholar]
- 8. Stevens V, Dumyati G, Fine LS, et al. . Cumulative antibiotic exposures over time and the risk of Clostridium difficile infection. Clin Infect Dis 2011; 53:42–8. [DOI] [PubMed] [Google Scholar]
- 9. Keller SC, Williams D, Gavgani M, et al. . Rates of and risk factors for adverse drug events in outpatient parenteral antimicrobial therapy. Clin Infect Dis 2018; 66:11–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Tamma PD, Avdic E, Li DX, et al. . Association of adverse events with antibiotic use in hospitalized patients. JAMA Intern Med 2017; 177:1308–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Society for Healthcare Epidemiology of America, Infecitous Diseases Society of America, Pediatric Infectious Diseases Society. Policy statement on antimicrobial stewardship by the Society for Healthcare Epidemiology of America (SHEA), the Infectious Diseases Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS). Infect Control Hosp Epidemiol 2012; 33:322–7. [DOI] [PubMed] [Google Scholar]
- 12. Daneman N, Rishu AH, Xiong W, et al. ; Canadian Critical Care Trials Group Duration of antimicrobial treatment for bacteremia in Canadian critically ill patients. Crit Care Med 2016; 44:256–64. [DOI] [PubMed] [Google Scholar]
- 13. Nelson AN, Justo JA, Bookstaver PB, et al. . Optimal duration of antimicrobial therapy for uncomplicated Gram-negative bloodstream infections. Infection 2017; 45:613–20. [DOI] [PubMed] [Google Scholar]
- 14. Heldman AW, Hartert TV, Ray SC, et al. . Oral antibiotic treatment of right-sided staphylococcal endocarditis in injection drug users: prospective randomized comparison with parenteral therapy. Am J Med 1996; 101:68–76. [DOI] [PubMed] [Google Scholar]
- 15. Mombelli G, Pezzoli R, Pinoja-Lutz G, et al. . Oral vs intravenous ciprofloxacin in the initial empirical management of severe pyelonephritis or complicated urinary tract infections: a prospective randomized clinical trial. Arch Intern Med 1999; 159:53–8. [DOI] [PubMed] [Google Scholar]
- 16. Talan DA, Stamm WE, Hooton TM, et al. . Comparison of ciprofloxacin (7 days) and trimethoprim-sulfamethoxazole (14 days) for acute uncomplicated pyelonephritis pyelonephritis in women: a randomized trial. JAMA 2000; 283:1583–90. [DOI] [PubMed] [Google Scholar]
- 17. San Pedro GS, Cammarata SK, Oliphant TH, Todisco T; Linezolid Community-Acquired Pneumonia Study Group Linezolid versus ceftriaxone/cefpodoxime in patients hospitalized for the treatment of Streptococcus pneumoniae pneumonia. Scand J Infect Dis 2002; 34:720–8. [DOI] [PubMed] [Google Scholar]
- 18. Goldberg E, Paul M, Talker O, et al. . Co-trimoxazole versus vancomycin for the treatment of methicillin-resistant Staphylococcus aureus bacteraemia: a retrospective cohort study. J Antimicrob Chemother 2010; 65:1779–83. [DOI] [PubMed] [Google Scholar]
- 19. Paul M, Bishara J, Yahav D, et al. . Trimethoprim-sulfamethoxazole versus vancomycin for severe infections caused by meticillin resistant Staphylococcus aureus: randomised controlled trial. BMJ 2015; 350:h2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Rodriguez-Pardo D, Pigrau C, Campany D, et al. . Effectiveness of sequential intravenous-to-oral antibiotic switch therapy in hospitalized patients with gram-positive infection: the SEQUENCE cohort study. Eur J Clin Microbiol Infect Dis 2016; 35:1269–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Zhao M, Liang L, Ji L, et al. . Similar efficacy and safety of daptomycin versus linezolid for treatment of vancomycin-resistant enterococcal bloodstream infections: a meta-analysis. Int J Antimicrob Agents 2016; 48:231–8. [DOI] [PubMed] [Google Scholar]
- 22. Bouza E, Diaz-Lopez MD, Bernaldo de Quiros JCL, Rodriguez-Creixems M. Ciprofloxacin in patients with bacteremic infections. Am J Med 1989; 87(Suppl 1):S228–31. [DOI] [PubMed] [Google Scholar]
- 23. Lily ZY, Jon DH. Outcomes of hospitalized neutropenic oncology patients with Pseudomonas aeruginosa bloodstream infections: focus on oral fluoroquinolone conversion. J Oncol Pharm Pract 2015; 22:584–90. [DOI] [PubMed] [Google Scholar]
- 24. Brigmon MM, Bookstaver PB, Kohn J, et al. . Impact of fluoroquinolone resistance in Gram-negative bloodstream infections on healthcare utilization. Clin Microbiol Infect 2015; 21:843–9. [DOI] [PubMed] [Google Scholar]
- 25. Morrill HJ, Morton JB, Caffrey AR, et al. . Antimicrobial resistance of Escherichia coli urinary isolates in the Veterans Affairs health care system. Antimicrob Agents Chemother 2017; 61:e02236-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Food and Drug Administration. FDA drug safety communication: FDA updates warnings for oral and injectable fluoroquinolone antibiotics due to disabling side effects. https://www.fda.gov/Drugs/DrugSafety/ucm511530.htm. Accessed 17 January 2018. [Google Scholar]
- 27. Nguyen AT, Gentry CA, Furrh RZ. A comparison of adverse drug reactions between high- and standard-dose trimethoprim-sulfamethoxazole in the ambulatory setting. Curr Drug Saf 2013; 8:114–9. [DOI] [PubMed] [Google Scholar]
- 28. Cheng MP, Bogoch II, Green K, et al. . Factors associated with 30-day mortality in respiratory infections caused by Streptococcus pneumoniae. Clin Infect Dis. 2018; 66:1282–5. [DOI] [PubMed] [Google Scholar]
- 29. Doi Y, Chambers H. Penicillins and beta-lactamase inhibitors. In: Bennett J, Dolin R, Blaser M. Mandell, Douglas, and Bennett’s Principles and Practices of Infections Diseases. 8th ed Philadelphia, PA: Elsevier Saunders; 2015:263–77. [Google Scholar]
- 30. Craig W, Andes D. Cephalosporins In: Bennett J, Dolin R, Blaser M. Mandell, Douglas, and Bennett’s Principles and Practices of Infectious Diseases. 8th ed Philadelphia, PA: Elsevier Saunders; 2015:278–92. [Google Scholar]
- 31. Siegel RE, Halpern NA, Almenoff PL, et al. . A prospective randomized study of inpatient IV. Antibiotics for community-acquired pneumonia. The optimal duration of therapy. Chest 1996; 110:965–71. [DOI] [PubMed] [Google Scholar]
- 32. File TM Jr, Segreti J, Dunbar L, et al. . A multicenter, randomized study comparing the efficacy and safety of intravenous and/or oral levofloxacin versus ceftriaxone and/or cefuroxime axetil in treatment of adults with community-acquired pneumonia. Antimicrob Agents Chemother 1997; 41:1965–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Ramirez JA, Bordon J. Early switch from intravenous to oral antibiotics in hospitalized patients with bacteremic community-acquired Streptococcus pneumoniae pneumonia. Arch Intern Med 2001; 161:848–50. [DOI] [PubMed] [Google Scholar]
- 34. Oosterheert JJ, Bonten MJ, Schneider MM, et al. . Effectiveness of early switch from intravenous to oral antibiotics in severe community acquired pneumonia: multicentre randomised trial. BMJ 2006; 333:1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. el Moussaoui R, de Borgie CA, van den Broek P, et al. . Effectiveness of discontinuing antibiotic treatment after three days versus eight days in mild to moderate-severe community acquired pneumonia: randomised, double blind study. BMJ 2006; 332:1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Postma DF, van Werkhoven CH, van Elden LJ, et al. ; CAP-START Study Group Antibiotic treatment strategies for community-acquired pneumonia in adults. N Engl J Med 2015; 372:1312–23. [DOI] [PubMed] [Google Scholar]
- 37. Viasus D, Simonetti AF, Garcia-Vidal C, et al. . Impact of antibiotic de-escalation on clinical outcomes in community-acquired pneumococcal pneumonia. J Antimicrob Chemother 2017; 72:547–53. [DOI] [PubMed] [Google Scholar]
- 38. Eriksson S, Zbornik J, Dahnsjö H, et al. . The combination of pivampicillin and pivmecillinam versus pivampicillin alone in the treatment of acute pyelonephritis. Scand J Infect Dis 1986; 18:431–8. [DOI] [PubMed] [Google Scholar]
- 39. Sandberg T, Englund G, Lincoln K, Nilsson LG. Randomised double-blind study of norfloxacin and cefadroxil in the treatment of acute pyelonephritis. Eur J Clin Microbiol Infect Dis 1990; 9:317–23. [DOI] [PubMed] [Google Scholar]
- 40. Johnson JR, Lyons MF 2nd, Pearce W, et al. . Therapy for women hospitalized with acute pyelonephritis: a randomized trial of ampicillin versus trimethoprim-sulfamethoxazole for 14 days. J Infect Dis 1991; 163:325–30. [DOI] [PubMed] [Google Scholar]
- 41. Cronberg S, Banke S, Bruno AM, et al. . Ampicillin plus mecillinam vs cefotaxime/cefadroxil treatment of patients with severe pneumonia or pyelonephritis: a double-blind multicentre study evaluated by intention-to-treat analysis. Scand J Infect Dis 1995; 27:463–8. [DOI] [PubMed] [Google Scholar]
- 42. Sandberg T, Alestig K, Eilard T, et al. . Aminoglycosides do not improve the efficacy of cephalosporins for treatment of acute pyelonephritis in women. Scand J Infect Dis 1997; 29:175–9. [DOI] [PubMed] [Google Scholar]
- 43. Moreno-Martínez A, Mensa J, Martínez JA, et al. . Cefixime versus amoxicillin plus netilmicin in the treatment of community-acquired non-complicated acute pyelonephritis. Med Clin (Barc) 1998; 111:521–4. [PubMed] [Google Scholar]
- 44. Sanchez M, Collvinent B, Miró O, et al. . Short-term effectiveness of ceftriaxone single dose in the initial treatment of acute uncomplicated pyelonephritis in women. A randomised controlled trial. Emerg Med J 2002; 19:19–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Monmaturapoj T, Montakantikul P, Mootsikapun P, Tragulpiankit P. A prospective, randomized, double dummy, placebo-controlled trial of oral cefditoren pivoxil 400mg once daily as switch therapy after intravenous ceftriaxone in the treatment of acute pyelonephritis. Int J Infect Dis 2012; 16:e843–9. [DOI] [PubMed] [Google Scholar]
- 46. Jernelius H, Zbornik J, Bauer CA. One or three weeks’ treatment of acute pyelonephritis? A double-blind comparison, using a fixed combination of pivampicillin plus pivmecillinam. Acta Med Scand 1988; 223:469–77. [DOI] [PubMed] [Google Scholar]
- 47. Mercuro NJ, Stogsdill P, Wungwattana M. A retrospective analysis comparing oral stepdown therapy for enterobacteriaceae bloodstream infections: fluoroquinolones versus beta-lactams. Int J Antimicrob Agents. 2018; 51:687–92. [DOI] [PubMed] [Google Scholar]
- 48. Kutob LF, Justo JA, Bookstaver PB, et al. . Effectiveness of oral antibiotics for definitive therapy of Gram-negative bloodstream infections. Int J Antimicrob Agents 2016; 48:498–503. [DOI] [PubMed] [Google Scholar]
- 49. Rieger KL, Bosso JA, MacVane SH, et al. . Intravenous-only or intravenous transitioned to oral antimicrobials for enterobacteriaceae-associated bacteremic urinary tract infection. Pharmacotherapy 2017; 37:1479–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Chastre J, Wolff M, Fagon JY, et al. ; PneumA Trial Group Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA 2003; 290:2588–98. [DOI] [PubMed] [Google Scholar]
- 51. Riccio LM, Popovsky KA, Hranjec T, et al. . Association of excessive duration of antibiotic therapy for intra-abdominal infection with subsequent extra-abdominal infection and death: a study of 2552 consecutive infections. Surg Infect (Larchmt) 2014; 15:417–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Spellberg B. The new antibiotic mantra—“shorter is better”. JAMA Intern Med 2016; 176:1254–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Mazuski JE, Tessier JM, May AK, et al. . The Surgical Infection Society revised guidelines on the management of intra-abdominal infection. Surg Infect (Larchmt) 2017; 18:1–76. [DOI] [PubMed] [Google Scholar]
- 54. Gomi H, Solomkin JS, Schlossberg D, et al. . Tokyo guidelines 2018: antimicrobial therapy for acute cholangitis and cholecystitis. J Hepatobiliary Pancreat Sci. 2018; 25:3–16. [DOI] [PubMed] [Google Scholar]
- 55. Havey TC, Fowler RA, Daneman N. Duration of antibiotic therapy for bacteremia: a systematic review and meta-analysis. Crit Care 2011; 15:R267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Christensen KL, Holman RC, Steiner CA, et al. . Infectious disease hospitalizations in the United States. Clin Infect Dis 2009; 49:1025–35. [DOI] [PubMed] [Google Scholar]
- 57. Eliakim-Raz N, Yahav D, Paul M, Leibovici L. Duration of antibiotic treatment for acute pyelonephritis and septic urinary tract infection– 7 days or less versus longer treatment: systematic review and meta-analysis of randomized controlled trials. J Antimicrob Chemother 2013; 68:2183–91. [DOI] [PubMed] [Google Scholar]
- 58. Kalil AC, Metersky ML, Klompas M, et al. . Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis 2016; 63:e61–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Wintenberger C, Guery B, Bonnet E, et al. . Recommendation Group of the SPILF Proposal for shorter antibiotic therapies. Med Mal Infect 2017; 47:92–141. [DOI] [PubMed] [Google Scholar]
- 60. Pugh R, Grant C, Cooke RP, Dempsey G. Short-course versus prolonged-course antibiotic therapy for hospital-acquired pneumonia in critically ill adults. Cochrane Database Syst Rev 2015; 8:CD007577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kilburn SA, Featherstone P, Higgins B, Brindle R. Interventions for cellulitis and erysipelas. Cochrane Database Syst Rev 2010; 6:CD004299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Li JZ, Winston LG, Moore DH, Bent S. Efficacy of short-course antibiotic regimens for community-acquired pneumonia: a meta-analysis. Am J Med 2007; 120:783–90. [DOI] [PubMed] [Google Scholar]
- 63. Klausner HA, Brown P, Peterson J, et al. . A trial of levofloxacin 750 mg once daily for 5 days versus ciprofloxacin 400 mg and/or 500 mg twice daily for 10 days in the treatment of acute pyelonephritis. Curr Med Res Opin 2007; 23:2637–45. [DOI] [PubMed] [Google Scholar]
- 64. Peterson J, Kaul S, Khashab M, et al. . A double-blind, randomized comparison of levofloxacin 750 mg once-daily for five days with ciprofloxacin 400/500 mg twice-daily for 10 days for the treatment of complicated urinary tract infections and acute pyelonephritis. Urology 2008; 71:17–22. [DOI] [PubMed] [Google Scholar]
- 65. Sandberg T, Skoog G, Hermansson AB, et al. . Ciprofloxacin for 7 days versus 14 days in women with acute pyelonephritis: a randomised, open-label and double-blind, placebo-controlled, non-inferiority trial. Lancet 2012; 380:484–90. [DOI] [PubMed] [Google Scholar]
- 66. van Nieuwkoop C, van der Starre WE, Stalenhoef JE, et al. . Treatment duration of febrile urinary tract infection: a pragmatic randomized, double-blind, placebo-controlled non-inferiority trial in men and women. BMC Med 2017; 15:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Léophonte P, Choutet P, Gaillat J, et al. . Efficacy of a ten day course of ceftriaxone compared to a shortened five day course in the treatment of community-acquired pneumonia in hospitalized patients with risk factors. Med Mal Infect 2002; 32:369–81. [Google Scholar]
- 68. Dunbar LM, Wunderink RG, Habib MP, et al. . High-dose, short-course levofloxacin for community-acquired pneumonia: a new treatment paradigm. Clin Infect Dis 2003; 37:752–60. [DOI] [PubMed] [Google Scholar]
- 69. Tellier G, Niederman MS, Nusrat R, et al. . Clinical and bacteriological efficacy and safety of 5 and 7 day regimens of telithromycin once daily compared with a 10 day regimen of clarithromycin twice daily in patients with mild to moderate community-acquired pneumonia. J Antimicrob Chemother 2004; 54:515–23. [DOI] [PubMed] [Google Scholar]
- 70. Léophonte P, File T, Feldman C. Gemifloxacin once daily for 7 days compared to amoxicillin/clavulanic acid thrice daily for 10 days for the treatment of community-acquired pneumonia of suspected pneumococcal origin. Respir Med 2004; 98:708–20. [DOI] [PubMed] [Google Scholar]
- 71. Chotiprasitsakul D, Han JH, Cosgrove SE, et al. ; Antibacterial Resistance Leadership Group Comparing the outcomes of adults with enterobacteriaceae bacteremia receiving short-course versus prolonged-course antibiotic therapy in a multicenter, propensity score-matched cohort. Clin Infect Dis 2018; 66:172–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Doi A, Morimoto T, Iwata K. Shorter duration of antibiotic treatment for acute bacteraemic cholangitis with successful biliary drainage: a retrospective cohort study. Clin Microbiol Infect. In press. [DOI] [PubMed] [Google Scholar]
- 73. Al-Hasan MN, Albrecht H, Bookstaver PB, et al. . Duration of antimicrobial therapy for Enterobacteriaceae bacteremia: using convenient end points for convenient conclusions. Clin Infect Dis. In press. [DOI] [PubMed] [Google Scholar]
- 74. Chotiprasitakul D, Han JH, Cosgrove SE, et al. . Reply to authors. Clin Infect Dis. In press. [Google Scholar]
- 75. van Lent AU, Bartelsman JF, Tytgat GN, et al. . Duration of antibiotic therapy for cholangitis after successful endoscopic drainage of the biliary tract. Gastrointest Endosc 2002; 55:518–22. [DOI] [PubMed] [Google Scholar]
- 76. Corona A, Wilson AP, Grassi M, Singer M. Prospective audit of bacteraemia management in a university hospital ICU using a general strategy of short-course monotherapy. J Antimicrob Chemother 2004; 54:809–17. [DOI] [PubMed] [Google Scholar]
- 77. Hedrick TL, McElearney ST, Smith RL, et al. . Duration of antibiotic therapy for ventilator-associated pneumonia caused by non-fermentative gram-negative bacilli. Surg Infect (Larchmt) 2007; 8:589–97. [DOI] [PubMed] [Google Scholar]
- 78. Kogure H, Tsujino T, Yamamoto K, et al. . Fever-based antibiotic therapy for acute cholangitis following successful endoscopic biliary drainage. J Gastroenterol 2011; 46:1411–7. [DOI] [PubMed] [Google Scholar]
- 79. Uno S, Hase R, Kobayashi M, et al. . Short-course antimicrobial treatment for acute cholangitis with Gram-negative bacillary bacteremia. Int J Infect Dis 2017; 55:81–5. [DOI] [PubMed] [Google Scholar]
- 80. Wagenlehner FM, Umeh O, Steenbergen J, et al. . 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 2015; 385:1949–56. [DOI] [PubMed] [Google Scholar]
- 81. Gil-Bermejo JM. Antibiotic treatment duration (7 vs 14 Days) comparison in blood stream infection causes by enterobacteriaceae (SHORTEN). https://clinicaltrials.gov/ct2/show/NCT02400268. Accessed 17 January 2018. [Google Scholar]
- 82. Daneman N. BALANCE on the wards: a pilot RCT (BALANCE-WARDS). https://clinicaltrials.gov/ct2/show/NCT02917551. Accessed 17 January 2018. [Google Scholar]
- 83. Yahav D. Duration of antibiotics for the treatment of Gram-negative bacilli bacteremia. https://clinicaltrials.gov/ct2/show/NCT01737320. Accessed 17 January 2018. [Google Scholar]
- 84. Huttner A. Antibiotic durations for Gram-negative bacteremia (PIRATE). https://clinicaltrials.gov/ct2/show/NCT03101072. Accessed 17 January 2018. [Google Scholar]
- 85. Daneman N. Bacteremia antibiotic length actually needed for clinical effectiveness (BALANCE). https://clinicaltrials.gov/ct2/show/NCT03005145. Accessed 17 January 2018. [Google Scholar]
- 86. Wiggers JB, Xiong W, Daneman N. Sending repeat cultures: is there a role in the management of bacteremic episodes? (SCRIBE study). BMC Infect Dis 2016; 16:286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Canzoneri CN, Akhavan BJ, Tosur Z, et al. . Follow-up blood cultures in Gram-negative bacteremia: are they needed?Clin Infect Dis 2017; 65:1776–9. [DOI] [PubMed] [Google Scholar]
- 88. Sayood S, Sutton J, Baures T, Spivak E. The utility of repeat blood cultures for bacteremic urinary tract infections and associated durations of therapy. Open Forum Infect Dis 2017; 4:S344–5. [Google Scholar]
- 89. Bordón J, Peyrani P, Brock GN, et al. ; CAPO Study Group The presence of pneumococcal bacteremia does not influence clinical outcomes in patients with community-acquired pneumonia: results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort study. Chest 2008; 133:618–24. [DOI] [PubMed] [Google Scholar]
- 90. Campbell SG, Marrie TJ, Anstey R, et al. . The contribution of blood cultures to the clinical management of adult patients admitted to the hospital with community-acquired pneumonia: a prospective observational study. Chest 2003; 123:1142–50. [DOI] [PubMed] [Google Scholar]
- 91. Chen CY, Chen YH, Lu PL, et al. . Proteus mirabilis urinary tract infection and bacteremia: risk factors, clinical presentation, and outcomes. J Microbiol Immunol Infect 2012; 45:228–36. [DOI] [PubMed] [Google Scholar]