Skip to main content
Log in

Ceftriaxone

An Update of its Use in the Management of Community-Acquired and Nosocomial Infections

  • Adis Drug Evaluation
  • Published:
Drugs Aims and scope Submit manuscript

Summary

Abstract

Ceftriaxone is a parenteral third-generation cephalosporin with a long elimination half-life which permits once-daily administration. It has good activity against Streptococcus pneumoniae, methicillin-susceptible staphylococci, Haemophilus influenzae, Moraxella catarrhalis and Neisseria spp. Although active against Enterobacteriaceae, the recent spread of derepressed mutants which hyperproduce chromosomal β-lactamases and extended-spectrum β-lactamases has diminished the activity of all third-generation cephalosporins against these pathogens necessitating careful attention to sensitivity studies.

Extensive data from randomised clinical trials confirm the efficacy of ceftriaxone in serious and difficult-to-treat community-acquired infections including meningitis, pneumonia and nonresponsive acute otitis media. Ceftriaxone also has efficacy in other community-acquired infections including uncomplicated gonorrhoea, acute pyelonephritis and various infections in children. In the nosocomial setting, extensive data also confirm the efficacy of ceftriaxone with or without an aminoglycoside in serious Gram-negative infections, pneumonia, spontaneous bacterial peritonitis and as surgical prophylaxis. Outpatient use of ceftriaxone, either as part of a step-down regimen or parenterally, is a distinguishing feature of the data gathered on the agent over the last decade. The review focuses on new applications of the drug and its use in infections in which the causative pathogens or their resistance patterns have changed over the past decade.

Ceftriaxone has a good tolerability profile, the most common events being diarrhoea, nausea, vomiting, candidiasis and rash. Ceftriaxone may cause reversible biliary pseudolithiasis, notably at higher dosages of the drug (≥2 g/day); however, the incidence of true lithiasis is <0.1%. Injection site discomfort or phlebitis can occur after intramuscular or intravenous administration.

Conclusions: As a result of its strong activity against S. pneumoniae, ceftriaxone holds an important place, either alone or as part of a combination regimen, in the treatment of invasive pneumococcal infections, including those with reduced β-lactam susceptibility. Its once-daily administration schedule allows simplification of otherwise complex regimens in a hospital setting and has also contributed to its popularity as a parenteral agent in an ambulatory setting. These properties, together with a well characterised tolerability profile, mean that ceftriaxone is likely to retain its place as an important third-generation cephalosporin in the treatment of serious community-acquired and nosocomial infections.

Antibacterial Activity

According to data from large in vitro studies published since 1998, ceftriaxone has good activity against Streptococcus pneumoniae, β-haemolytic group streptococci and methicillin-susceptible staphylococci. Its activity is more variable against viridans group streptococci and, as for other β-lactam agents, ceftriaxone is inactive against methicillin-resistant staphylococci and enterococci. Although penicillin-resistant S. pneumoniae are becoming increasingly common, the prevalence of resistance to ceftriaxone among S. pneumoniae remains <5% worldwide.

Ceftriaxone is highly active against Haemophilus influenzae, Moraxella catarrhalis, Neisseria spp. and has good activity against Salmonella and Shigella spp. However, data published since 1998 show that ceftriaxone, like other third-generation cephalosporins, now has variable activity against most Enterobacteriaceae. Ceftriaxone has little or no activity against Acinetobacter spp., Pseudomonas aeruginosa, Stenotrophomonas maltophilia or Achromobacter xylosoxidans.

Ceftriaxone is stable against TEM-1, TEM-2, SHV-1 and K1 chromosomal β-lactamases. Its activity is also unaffected by inducible AmpC β-lactamases. However, like other third-generation agents, its activity is reduced by derepressed mutants which hyperproduce chromosomal β-lactamases or extended-spectrum β-lactamases, both of which are now widespread. Other relevant mechanisms of resistance are modification or acquisition of supplementary penicillin-binding proteins, usually in Gram-positive bacteria. Resistance attributable to reduced porin expression or efflux is less common.

Studies in patients show that ceftriaxone reversibly alters the intestinal flora (suppresses enterobacteria, bifidobacteria, clostridia and Bacteroides and increases enterococci and Candida), but is associated with the emergence of fewer cephalosporin-resistant Gram-negative bacilli than cefotaxime or cefazolin in a randomised study. Additionally, in a study in 12 patients, the colonic microflora was found to normalise within 14 days of completion of treatment in all but one recipient.

The inter-relationship between pharmacodynamic and pharmacokinetic parameters is also important in predicting antibacterial activity. A pharmacodynamic review of the Alexander Project surveillance data indicated that ceftriaxone was associated with plasma concentrations above the minimum inhibitory concentration required to inhibit the growth of 90% of isolates (MIC90) of S. pneumoniae, H. influenzae, M. catarrhalis and Staphylococcus aureus for >50% of the dosage interval. In healthy adult volunteers, serum concentrations of ceftriaxone (intravenous 30 minute infusion of 1g once daily) were bactericidal for 100% of the dosage interval against clinical isolates of S. pneumoniae, Klebsiella pneumoniae, Enterobacter aerogenes, Escherichia coli, Serratia marcescens and H. influenzae. In an in vitro study evaluating penicillin-susceptible, -intermediate and -resistant S. pneumoniae clinical isolates, ceftriaxone exerted post antibiotic effects (PAEs) ranging from 1 to 7.2 hours after isolates had been exposed to concentrations 10 times the MIC value. In another study, mean PAEs against S. aureus, S. pneumoniae, H. influenzae and E. coli strains were 0.9, 2.6, −0.8 and −2.1 hours, respectively, reflecting the fact that cephalosporins generally only demonstrate a PAE against Gram-positive bacteria.

Pharmacokinetic Profile

Intravenous ceftriaxone 0.5, 1 or 2g produces mean peak plasma concentrations (Cmax) of 82, 151 and 257 mg/L, respectively, whereas intramuscular ceftriaxone 0.5 or 1g achieves Cmax values of 38 and 76 mg/L, respectively, after 2 to 3 hours. 24 hours after intravenous ceftriaxone 2g, mean plasma concentrations range from 12 to 20 mg/L.

Repeated once-daily intravenous administration of ceftriaxone 2g results in an 8% increase in mean Cmax, and repeated intramuscular administration of ceftriaxone 1g results in 11% accumulation of the drug.

Ceftriaxone binds reversibly to albumin and the level of binding decreases with increasing ceftriaxone plasma concentrations (≈95% at >70 mg/L to ≈58% at 600 mg/L).

Ceftriaxone distributes widely in the body fluids and tissues. The volume of distribution of ceftriaxone in healthy volunteers ranges from 5.8 to 15.5L.

Ceftriaxone preferentially localises in bile and mean concentrations of ≈153 and ≈44 mg/L are obtained 1 and ≈3 hours, respectively, after intravenous administration of lg dose. The drug is primarily eliminated unchanged by the kidneys; 45 to 60% of a 0.5 to 3g dose is excreted in the urine of healthy subjects within 48 hours. The remainder is secreted in the bile and the faeces as microbiologically inactive compounds.

Total plasma clearance of ceftriaxone is dose related; it increases from mean values of 0.61 to 1.0 L/h after a 0.5g intravenous dose to 1.18 and 1.29 L/h after a 2g intravenous dose.

The mean elimination half-life (t½) of ceftriaxone in healthy adults is ≈6 to 9 hours, which is considerably longer than that of other cephalosporins (0.6 to 4.4 hours). The t½ of ceftriaxone does not vary according to dose size, frequency or route of administration.

Therapeutic Use in Community-Acquired Infections

Meningitis: In five randomised trials, intravenous ceftriaxone 60 to 100 mg/kg sterilised the CSF in 98 to 100% of infants and children with meningitis within 18 to 36 hours. Similar response rates were observed with alatrofloxacin, cefuroxime and cefotaxime (88 to 97% of patients), although cefuroxime was associated with delayed sterilisation more frequently than ceftriaxone (12 vs 2%). Neurological sequelae at discharge were reported in 9 to 23% of patients who received ceftriaxone. Two small comparative studies also showed that ceftriaxone was effective in the treatment of adults with meningitis.

Acute otitis media: According to data from six randomised trials, a single intramuscular dose of ceftriaxone 50 mg/kg had similar clinical efficacy to 10-day oral regimens of amoxicillin plus or minus clavulanic acid, cefaclor or cotrimoxazole (trimethoprim/sulfamethoxazole) in infants and children with uncomplicated acute otitis media (AOM). In children with AOM unresponsive to previous antibacterial therapy, three trials showed that a three-dose regimen of intramuscular ceftriaxone 50 mg/kg/day eradicated 93 to 98% of middle ear pathogens. Significantly lower eradication rates were observed with one versus three doses of ceftriaxone in this patient group in one comparative trial (73 vs 98%; p < 0.001).

A flexible approach advocated by McCracken recommends that in uncomplicated AOM a single dose of ceftriaxone be given, with additional doses if necessary (after 48 to 72 hours) if there is a clinical failure with the first dose. It is suggested that the second dose may be given at 48 to 72 hours rather than 24 hours, since the concentration of ceftriaxone exceeds the MIC90 for approximately 120 hours or longer for most organisms including penicillin-intermediate and -resistant strains of S. pneumoniae. This approach allows for the possibility that some children failing the one-dose ceftriaxone regimen may be cured by the second dose and hence not require the third dose.

Community-acquired pneumonia: According to data from ten randomised trials in hospitalised adults with community-acquired pneumonia (CAP), ceftriaxone 1 to 4 g/day, alone or as part of a step-down regimen, had similar efficacy to that of other β-lactam agents (amoxicillin/clavulanic acid, cefepime, cefpodoxime proxetil, cefodizime), fluoroquinolones (ciprofloxacin, levofloxacin, gatifloxacin, trovafloxacin) and linezolid. Clinical success rates with ceftriaxone-based therapy ranged from 81 to 98% versus 85 to 98% with comparator regimens. A step-down regimen of ceftriaxone 50 mg/kg/day followed by oral cefixime or amoxicillin/clavulanic acid achieved similar clinical success rates in children with CAP in a randomised trial.

Additionally, in a nonblind study in 31 patients with CAP, the average length of stay in hospital was reduced by 1.2 days (4.8 vs 6 days) in patients receiving step-down therapy with intravenous ceftriaxone (dosage not reported) followed by oral cefpodoxime proxetil 200mg twice daily compared with those receiving ceftriaxone followed by other oral treatment regimens (choice of oral agent was at the physician’s discretion). There were no recurrences of primary infection or readmissions to hospital at 1-month follow-up in either treatment group.

Invasive infections caused by ceftriaxone-nonsusceptible Streptococcus pneumoniae: In patients with pneumococcal meningitis, treatment failures or unpredictable responses have been observed with strains with MICs ≥1 mg/L. Combination therapy with vancomycin or rifampicin is therefore recommended for patients with ceftriaxone-nonsusceptible strains, although published data concerning the efficacy of these regimens remain very limited. In contrast, recent data in other pneumococcal infections (predominantly pneumonia and bacteraemia) suggest that ceftriaxone can be used effectively to treat infections caused by S. pneumoniae with ceftriaxone MICs ≤2 mg/L.

Sexually transmitted infections: Single dose intramuscular ceftriaxone 0.25g consistently produces bacteriological eradication rates of ≥95% in adults with uncomplicated gonorrhoea according to 15 large randomised trials. Eradication rates achieved with ceftriaxone were similar to those reported with single doses of comparator agents (90 to 100%; cefixime, ceftizoxime, cefotaxime, ampicillin/sulbactam plus probenecid, trospectomycin, ciprofloxacin, fleroxacin and enoxacin).

A single intramuscular dose of ceftriaxone 0.25g combined with oral doxycycline achieved clinical success in 95% of women with mild to moderate pelvic inflammatory disease compared with 97% treated with oral clindamycin plus ciprofloxacin in a randomised trial.

Acute pyelonephritis: In pregnant women with acute pyelonephritis, four randomised trials showed that parenteral ceftriaxone 1 g/day followed by oral cefalexin or cefradine achieved bacteriological cure in 92 and 98% of patients. Treatment was effective whether administered completely or partially on an outpatient basis, although medical or infectious complications precluded outpatient management in over 50% of women at >24 weeks gestation. Similar results were achieved with comparator regimens (intravenous cefazolin or ampicillin plus gentamicin followed by similar step-down therapy). No differences in birth outcomes were noted between regimens.

Other paediatric infections: Data from randomised trials also show that ceftriaxone is effective in the treatment of acute pyelonephritis, multidrug-resistant Salmonella typhi septicaemia, typhoid fever, skin and soft-tissue infections, bone infections, infectious diarrhoea, febrile episodes in children with sickle haemoglobinopathies and in the prevention of invasive infections in febrile children (aged 3 to 36 months). Furthermore, regimens including ceftriaxone have proven effective and were generally well tolerated in neonates (including newborn) with proven or suspected infection.

Therapeutic Use in Nosocomial Infections

Gram-negative infections: One large multinational randomised trial showed that ceftriaxone plus tobramycin achieved similar clinical response rates to ceftazidime monotherapy in adult hospitalised patients with serious nosocomial Gram-negative infections (59 to 76% vs 73 to 80% depending on the infection site).

Pneumonia: In patients with nosocomial pneumonia, intravenous ceftriaxone 2 g/day achieved clinical success in 70% of patients compared with 80% of patients treated with intravenous cefoperazone in a randomised trial.

Surgical or procedural prophylaxis: Ceftriaxone was associated with wound infection rates of 3 to 6% after abdominal or biliary surgery, 3% after breast or gynaecological surgery, 0.5% after orthopaedic surgery and 15% after percutaneous endoscopic gastrotomy. In all studies, ceftriaxone was administered intravenously as a single 1 or 2g dose prior to or during surgery. The rate of postoperative wound infections observed with ceftriaxone was generally similar to that achieved with comparator regimens, except for two studies in which ceftriaxone was associated with a significantly lower infection rate: 0.5 vs 4% with placebo in orthopaedic surgery (p < 0.001) and 6 vs 17% with metronidazole in combination with gentamicin in bowel surgery (p < 0.05).

Spontaneous bacterial peritonitis: In two small prospective trials, empirical treatment with intravenous ceftriaxone 2 g/day achieved a bacteriological cure in 95 and 100% of patients with spontaneous bacterial peritonitis. This compared with a bacteriological cure rate of 94% with intravenous cefonicid in a randomised trial.

Tolerability

In general, adverse events associated with ceftriaxone were mild to moderate in severity and resolved spontaneously. With rare exceptions, severe or serious events were not observed after single dose treatment with ceftriaxone in adults, children or infants. In addition, studies in neonates also indicated that ceftriaxone monotherapy or combination therapy was generally well tolerated.

According to data from clinical trials, gastrointestinal events are the most common adverse events associated with ceftriaxone. In general, diarrhoea was reported in 1 to 15% of patients, nausea and vomiting in ≤4%, and abdominal pain or discomfort in ≤2% of patients. All third-generation cephalosporins, including ceftriaxone, have been associated with rare reports of pseudomembranous colitis; therefore, overgrowth of Clostridium difficile should be considered in patients with diarrhoea. Other systemic events associated with ceftriaxone included rash in ≤6% of patients, candidiasis (oral or vaginal) in about 4 to 5% of patients and pruritus, headache and dizziness in ≤3% of patients each.

Ceftriaxone can cause reversible biliary pseudolithiasis, usually at higher dosages (≥2 g/day); however, the incidence of true lithiasis is <0.1%. Data from six small incidence studies (n = 8 to 118 patients in each study) show that sonographic abnormalities indicative of pseudolithiasis were observed in 12 to 43% (n = 37 patients in the latter study) of adolescents and children and 21 and 25% of adults treated with intravenous ceftriaxone. Up to one-third (0 to 33%) of patients developed symptoms, but pseudolithiasis resolved spontaneously in all cases.

Haematological reactions with ceftriaxone include anaemia, haemolytic anaemia, leucopenia, neutropenia, thrombocytopenia, eosinophilia, agranulocytosis and a positive Coomb’s test. Prolongation of prothrombin time has been observed rarely with ceftriaxone. As with other third-generation agents, mild reversible abnormalities in liver function tests have been documented with ceftriaxone. Ceftriaxone appears to have a low nephrotoxic potential and can be coad-ministered with aminoglycosides.

Ceftriaxone can cause discomfort or pain at the injection site following intramuscular administration, and phlebitis following intravenous injection. In clinical trials, the incidence of these events ranged from 0 to 45% and up to 11%, respectively. Of note, in comparison with water, 1% lidocaine has been shown to reduce the severity and duration of pain associated with ceftriaxone injection (250 to 1g) in healthy volunteers (aged 14 to 55 years) in randomised, single- or double-blind trials.

Comparative data with ceftriaxone monotherapy are limited because the drug is often administered as part of a step-down regimen or in combination with other agents. Available data show that the incidence of adverse events with ceftriaxone is generally similar to that of other β-lactams and fluoroquinolones in adults and children. Significant differences noted between regimens include fewer adverse reactions, notably gastrointestinal events, than amoxicillin/clavulanic acid and fewer gastrointestinal events than cefixime but higher incidences of diarrhoea than cotrimoxazole and cefpodoxime proxetil and rash compared with oral amoxicillin.

Dosage and Administration

Ceftriaxone can be administered by intravenous or intramuscular injection. The recommended dose in adults and children (aged ≥12 years) is 1 to 2g once daily (or in two equally divided doses) in the US or 1g once daily in the UK. The total daily dose should not exceed 4g. A single intramuscular dose of 250mg is recommended for the treatment of uncomplicated gonorrhoea. For surgical prophylaxis, a single intravenous 1g dose given 0.5 to 2 hours before surgery is advised.

In children aged <12 years, ceftriaxone 50 to 75 mg/kg/day is recommended for serious infections and a dose of 100 mg/kg/day is recommended for meningitis in the US; the dose may be administered once daily or split into two equally divided doses. In the UK, ceftriaxone 20 to 50 mg/kg once daily is recommended as the standard dose, with doses of up to 80 mg/kg for severe infections. AOM should be treated with a single intramuscular dose of 50 mg/kg.

Dosage adjustment is not required in patients with impaired hepatic or renal function, although plasma concentrations of ceftriaxone should be monitored regularly in patients with concomitant renal and hepatic dysfunction.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Table I
Table II
Table III
Fig. 2
Fig. 1
Table IV
Fig. 3
Table V
Table VI
Table VII
Table VIII
Table IX
Table X
Table XI
Fig. 4
Table XII
Table XIII
Table XIV

Similar content being viewed by others

References

  1. Richards DM, Heel RC, Brogden RN, et al. Ceftriaxone: a review of its antibacterial activity, pharmacological properties and therapeutic use. Drugs 1984; 27: 469–527

    Article  PubMed  CAS  Google Scholar 

  2. Brogden RN, Ward A. Ceftriaxone: a reappraisal of its antibacterial activity and pharmacokinetic properties, and an update on its therapeutic use with particular reference to once-daily administration. Drugs 1988 Jun; 35: 604–45

    Article  PubMed  CAS  Google Scholar 

  3. Jones RN, Pfaller MA, Doern GV, et al. Antimicrobial activity and spectrum investigation of eight broad-spectrum β-lactam drugs: a 1997 surveillance trial in 102 medical centers in the United States. Diagn Microbiol InfectDis 1998 Mar; 30: 215–28

    Article  CAS  Google Scholar 

  4. Diekema DJ, Coffman SL, Marshall SA, et al. Comparison of activities of broad-spectrum β-lactam compounds against 1,128 Gram-positive cocci recently isolated in cancer treatment centers. Antimicrob Agents Chemother 1999 Apr; 43: 940–3

    PubMed  CAS  Google Scholar 

  5. Thornsberry C, Jones ME, Hickey ML, et al. Resistance surveillance of Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis isolated in the United States, 1997–1998. J Antimicrob Chemother 1999 Dec; 44: 749–59

    Article  PubMed  CAS  Google Scholar 

  6. Jones RN, Pfaller MA, Doern GV. Comparative antimicrobial activity of trovafloxacin tested against 3049 Streptococcus pneumoniae isolates from the 1997–1998 respiratory infection season. Diagn Microbiol Infect Dis 1998 Oct; 32: 119–26

    Article  PubMed  CAS  Google Scholar 

  7. Doern GV, Jones RN, Pfaller MA, et al. Bacterial pathogens isolated from patients with skin and soft tissue infections: frequency of occurrence and antimicrobial susceptibility patterns from the SENTRY Antimicrobial Surveillance Program (United States and Canada, 1887). Diagn Microbiol InfectDis 1999 May; 34: 65–72

    Article  CAS  Google Scholar 

  8. Jones RN, Jenkins SG, Hoban DJ, et al. In vitro activity of selected cephalosporins and erythromycin against staphylococci and pneumococci isolated at 38 North American medical centers participating in the SENTRY Antimicrobial Surveillance Program, 1997–1998. Diagn Microbiol Infect Dis 2000 Jun; 37: 93–8

    Article  PubMed  CAS  Google Scholar 

  9. Low DE, de Azavedo J, Canadian Bacterial Surveillance Network. In vitro activity of cefepime against multidrug-resistant Gram-negative bacilli, viridans groups streptococci and Streptococcus pneumoniae from a cross-Canada surveillance study. Can J Infect Dis 1999 Mar–Apr; 10: 122–7

    PubMed  CAS  Google Scholar 

  10. Diekema DJ, Pfaller MA, Jones MA, et al. Trends in antimicrobial susceptibility of bacterial pathogens isolated from patients with bloodstream infections in the USA, Canada and Latin America. Int J Antimicrob Agents 2000; 13: 257–71

    Article  PubMed  CAS  Google Scholar 

  11. Pfaller MA, Jones RN, Doern GV, et al. Survey of blood stream infections attributable to gram-positive cocci: frequency of occurrence and antimicrobial susceptibility of isolates collected in 1997 in the United States, Canada, and Latin America from the SENTRY Antimicrobial Surveillance Program. Diagn Microbiol Infect Dis 1999; 33: 283–97

    Article  PubMed  CAS  Google Scholar 

  12. Diekema DJ, Pfaller MA, Jones RN, et al. Survey of bloodstream infections due to gram-negative bacilli: frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, and Latin America for the SENTRY Antimicrobial Surveillance Program, 1997. Clin Infect Dis 1999 Sep; 29: 595–607

    Article  PubMed  CAS  Google Scholar 

  13. Santos JI, Arredondo JL, Vazquez V. Bacterial isolates from hematology, oncology, and intensive care units: susceptibility to cefpirome and other beta-lactam antibiotics in a multicenter study in Mexico. Curr Ther Res Clin Exp 2000 May; 61: 266–76

    Article  CAS  Google Scholar 

  14. Fluit AC, Jones ME, Schmitz F-J, et al. Antimicrobial susceptibility and frequency of occurrence of clinical blood isolates in Europe from the SENTRY Antimicrobial Surveillance Program, 1997 and 1998. Clin Infect Dis 2000 Mar; 30: 454–60

    Article  PubMed  CAS  Google Scholar 

  15. Hanberger H, Garcia-Rodriguez J-A, Gobernado M, et al. Antibiotic susceptibility among aerobic gram-negative bacilli in intensive care units in 5 European countries. JAMA 1999 Jan 6; 281: 67–71

    Article  PubMed  CAS  Google Scholar 

  16. Baquero F, Garcia-Rodriguez JA, Garcia de Lomas J, et al. Antimicrobial resistance of 1,113 Streptococcus pneumoniae isolates from patients with respiratory tract infections in Spain: results of a 1-year (1996–1997) multicenter surveillance study. Antimicrob Agents Chemother 1999 Feb; 43: 357–9

    PubMed  CAS  Google Scholar 

  17. Felmingham D, Gruneberg RN, Alexander PG. The Alexander Project 1996–1997: latest susceptibility data from this international study of bacterial pathogens from community-acquired lower respiratory tract infections. J Antimicrob Chemother 2000 Feb; 45: 191–203

    Article  PubMed  CAS  Google Scholar 

  18. Sahm DF, Jones ME, Hickey ML, et al. Resistance surveillance of Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis isolated in Asia and Europe, 1997–1998. J Antimicrob Chemother 2000 Apr; 45: 457–66

    Article  PubMed  CAS  Google Scholar 

  19. Jones RN, Hare RS, Sabatelli FJ, et al. In vitro Gram-positive antimicrobial activity of evernimicin (SCH 27899), a novel oligosaccharide, compared with other antimicrobials: a multi-centre international trial. J Antimicrob Chemother 2001 Jan; 47: 15–25

    Article  PubMed  CAS  Google Scholar 

  20. Thornsberry C, Sahm DF. Antimicrobial resistance in respiratory tract pathogens: results of an international surveillance study. Chemotherapy Basel 2000; 46 Suppl. 1: 15–23

    Article  CAS  Google Scholar 

  21. Turnidge JD, Bell JM, Collignon PJ, et al. Rapidly emerging antimicrobial resistances in Streptococcus pneumoniae in Australia. Med J Aust 1999 Feb 15; 170: 152–5

    PubMed  CAS  Google Scholar 

  22. National Committee for Clinical Laboratory Standards. Performance standards for antimicrobial susceptibility testing: twelfth informational supplement. Wayne (PA): NCCLS, 2002. NCCLS document M100-S12

  23. Kaplan Jr SL, Mason EO, Barson WJ, et al. Outcome of invasive infections outside the central nervous system caused by Streptococcus pneumoniae isolates nonsusceptible to ceftriaxone in children treated with beta-lactam antibiotics. Pediatr Infect Dis J 2001 Apr; 20(4): 392–6

    Article  PubMed  CAS  Google Scholar 

  24. Klugman KP, Feldman C. Penicillin- and cephalosporin-resistant Streptococcus pneumoniae: emerging treatment for an emerging problem. Drugs 1999 Jul; 58: 1–4

    Article  PubMed  CAS  Google Scholar 

  25. American Academy of Pediatrics: Committee on Infectious Diseases. Therapy for children with invasive pneumococcal infections. Pediatrics 1997 Feb; 99(2): 289–99

    Article  Google Scholar 

  26. Bartlett JG, Dowell SF, Mandell LA, et al. Practice guidelines for the management of community-acquired pneumonia in adults. Clin Infect Dis 2000 Aug; 31: 347–82

    Article  PubMed  CAS  Google Scholar 

  27. Sahm DF, Thornsberry C, Mayfield DC, et al. In vitro activities of broad-spectrum cephalosporins against nonmeningeal isolates of Streptococcus pneumoniae: MIC interpretation using NCCLS M100-S12 recommendations. J Clin Microbiol 2001; 40: 669–74

    Article  CAS  Google Scholar 

  28. Livermore DM. β-Lactamases in laboratory and clinical resistance. Clin Microbiol Rev 1995; 8(4): 557–84

    PubMed  CAS  Google Scholar 

  29. Bush K. Is it important to identify extended-spectrum beta-lactamase-producing isolates? Eur J Clin Microbiol Infect Dis 1996; 15(5): 361–4

    Article  PubMed  CAS  Google Scholar 

  30. National Committee for Clinical Laboratory Standards. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically; approved standard. 5th ed. v. 17. Wayne (PA): NCCLS, 2000. (NCCLS document M7-A5)

  31. Thornsberry C, Ogilvie PT, Holley HP, et al. Survey of susceptibilities of Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis isolates to 26 antimoicrobial agents: a prospective US study. Antimicrob Agents Chemother 1999; 43: 2612–23

    PubMed  CAS  Google Scholar 

  32. Johnson AP, Livermore DM, Woodford N, et al. High-level β-lactam resistance in strains of Streptococcus pneumoniae isolated in the UK [letter]. J Antimicrob Chemother 1998 Jul; 42: 115–6

    Article  PubMed  CAS  Google Scholar 

  33. Sloas MM, Barrett FF, Chesney PJ, et al. Cephalosporin treatment failure in penicillin- and cephalosporin-resistant Streptococcus pneumoniae meningitis. Pediatr Infect Dis J 1992; 11(8): 662–6

    PubMed  CAS  Google Scholar 

  34. Karlowsky JA, Jones ME, Mayfield DC, et al. In vitro evaluation of ceftriaxone activity against recent Gram-positive clinical isolates: results from the TSN database-US, 1996–2000 [abstract no. P1299]. Clin Microbiol Infect 2001; 7 Suppl. 1: 275

    Google Scholar 

  35. Unzaga MJ, Rojo P, Melero P, et al. Antibiotic susceptibility of 12 strains of Alloiococcus otitidis [abstract]. 37th Interscience Conference on Antimicrobial Agents and Chemotherapy 1997 Sep 28; 118

  36. Mayfield DC, Karlowsky JA, Sahm DF, et al. In vitro evaluation of ceftriaxone activity against recent Gram-negative clinical isolates: results from the TSN database-US, 1996–2000 [abstract no. P1300]. Clin Microbiol Infect 2001; 7 Suppl. 1: 275

    Google Scholar 

  37. Fuchs PC, Barry AL, Brown SD, et al. Survey of antimicrobial activity of four commonly used third generation cephalosporins tested against recent bacterial isolates from ten American medical centers, and assessment of disk diffusion test performance. Diagn Microbiol Infect Dis 1996 Apr; 24: 213–9

    Article  PubMed  CAS  Google Scholar 

  38. Herikstad H, Hayes PS, Hogan J, et al. Ceftriaxone-resistant Salmonella in the United States. Pediatr Infect Dis J 1997 Sep; 16: 904–5

    Article  PubMed  CAS  Google Scholar 

  39. Abadi FJR, Yakubu DE, Pennington TH. Antimicrobial susceptibility of penicillin-sensitive and penicillin-resistant meningococci. J Antimicrob Chemother 1995 May; 35: 687–90

    Article  PubMed  CAS  Google Scholar 

  40. Blondeau JM, Yaschuk Y In vitro activities of ciprofloxacin, cefotaxime, ceftriaxone, chloramphenicol, and rifampin against fully susceptible and moderately penicillin-resistant Neisseria meningitidis. Antimicrob Agents Chemother 1995 Nov; 39: 2577–9

    Article  PubMed  CAS  Google Scholar 

  41. Pascual A, Joyanes P, Martínez-Martínez L, et al. Comparison of broth microdilution and E-test for susceptibility testing of Neisseria meningitidis. J Clin Microbiol 1996 Mar; 34: 588–91

    PubMed  CAS  Google Scholar 

  42. Marshall SA, Rhomberg PR, Jones RN. Comparative evaluation of Etest for susceptibility testing Neisseria meningitidis with eight antimicrobial agents: an investigation using U.S. food and drug administration regulatory criteria. Diagn Microbiol Infect Dis 1997 Mar; 27: 93–7

    Article  PubMed  CAS  Google Scholar 

  43. van de Beek D, Hensen EF, Spanjaard L, et al. Meropenem susceptibility of Neisseria meningitidis and Streptococcus pneumoniae from meningitis patients in The Netherlands. J Antimicrob Chemother 1997 Dec; 40: 895–7

    Article  PubMed  Google Scholar 

  44. Schwebke JR, Whittington W, Rice RJ, et al. Trends in susceptibility of Neisseria gonorrhoeae to ceftriaxone from 1985 through 1991. Antimicrob Agents Chemother 1995 Apr; 39: 917–20

    Article  PubMed  CAS  Google Scholar 

  45. Tansuphasiri U, Kabklang S, Wongba C. Comparative in vitro activities of nine antimicrobials against pencillinase producing (PPNG) and nonproducing (nonPPNG) Neisseria gonorrhoeae isolated from patients attending Bangrak hospital, Bangkok. J Infect Dis Antimicrob Agents 1998 Sep–Dec; 15: 129–37

    Google Scholar 

  46. Wenling C, Xibao Z, Shi F, et al. Analysis of the antibiotic sensitivity of Neisseria gonorrheae in Guangzhou, Peoples Republic of China. Sex Transm Dis 2000 Sep; 27: 480–2

    Article  PubMed  CAS  Google Scholar 

  47. Tanaka M, Nakayama H, Haraoka M, et al. Antimicrobial resistance of Neisseria gonorrhoeae and high prevalence of ciprofloxacin-resistant isolates in Japan, 1993 to 1998. J Clin Microbiol 2000 Feb; 38: 521–5

    PubMed  CAS  Google Scholar 

  48. Tapsall JW, Phillips EA. The sensitivity of 173 Sydney isolates of Neisseria gonorrhoeae to cefpodoxime and other antibiotics used to treat gonorrhea. Pathology 1995 Jan; 27: 64–6

    Article  PubMed  CAS  Google Scholar 

  49. Chevalier B, Crenn Y, Cavallo JD, et al. Comparative activity of azithromycin against 100 strains of Neisseria gonorrhoeae [in French]. Pathol Biol 1995 Apr; 43: 281–3

    PubMed  CAS  Google Scholar 

  50. Talbot H, Romanowski B. In vitro activities of sparfloxacin, ceftriaxone, penicillin, tetracycline and doxycycline against Chlamydia trachomatis and Neisseria gonorrhoeae. Can J Infect Dis 1992 May–Jun; 3: 114–7

    Google Scholar 

  51. Wexler HM, Molitoris D, Finegold SM. In vitro activities of MK-826 (L-749,345) against 363 strains of anaerobic bacteria. Antimicrob Agents Chemother 2000 Aug; 44: 2222–4

    Article  PubMed  CAS  Google Scholar 

  52. Hamilton-Miller JMT, Shah S. Comparative anti-anaerobic activity of Men 10700, a penem antibiotic. Int J Antimicrob Agents 1998 Nov; 10: 321–4

    Article  PubMed  CAS  Google Scholar 

  53. Livermore DM. β-Lactamase-mediated resistance and opportunities for its control. J Antimicrob Chemother 1998; 41 Suppl. D: 25–41

    Article  PubMed  CAS  Google Scholar 

  54. Moosdeen F. The evolution of resistance to cephalosporins. Clin Infect Dis 1997 Mar; 24: 487–93

    Article  PubMed  CAS  Google Scholar 

  55. Livermore DM. Are all β-lactams created equal? Scand J Infect Dis 1996 Suppl. 101: 33–43

  56. Pitout JDD, Sanders CC, Sanders Jr WE, et al. Antimicrobial resistance with focus on β-lactam resistance in gram-negative bacilli. Am J Med 1997 Jul; 103: 51–9

    Article  PubMed  CAS  Google Scholar 

  57. Sanders CC, Sanders Jr WE. β-Lactam resistance in Gram-negative bacteria: global trends and clinical impact. Clin Infect Dis 1992 Nov; 15: 824–39

    Article  PubMed  CAS  Google Scholar 

  58. Du Bois SK, Marriott MS, Amyes SGB. TEM- and SHV-derived extended-spectrum β-lactamases: relationship between selection, structure and function. J Antimicrob Chemother 1995; 35: 7–22

    Article  PubMed  Google Scholar 

  59. Coffey TJ, Daniels M, McDougal LK, et al. Genetic analysis of clinical isolates of Streptococcus pneumoniae with high-level resistance to expanded-spectrum cephalosporins. Antimicrob Agents Chemother 1995 Jun; 39: 1306–13

    Article  PubMed  CAS  Google Scholar 

  60. Cavallaro V, Catania V, Bonaccorso R, et al. Effect of a broad-spectrum cephalosporin on the oral and intestinal microflora in patients undergoing colorectal surgery. J Chemother 1992; 4(2): 82–7

    PubMed  CAS  Google Scholar 

  61. Vogel F, Boedn G, Nilsson-Ehle I, et al. Effect of step-down therapy of ceftriaxone plus loracarbef versus parenteral therapy of ceftriaxone on the intestinal microflora in patients with community-acquired pneumonia [abstract]. Clin Microbiol Infect 2000; 6 Suppl. 1: 137

    Google Scholar 

  62. Nilsson-Ehle I, Nord CE, Ursing B. Ceftriaxone: pharmacokinetics and effect on the intestinal flora in patients with acute bacterial infections. Scand J Infect Dis 1985; 17: 77–82

    Article  PubMed  CAS  Google Scholar 

  63. Laing F, Ramotar K, Krulicki W, et al. Comparison of cefotaxime and ceftriaxone on the induction of cephalosporin-resistant Gram-negative bacilli in the intestinal tract of hospital patients [abstract]. 35th Interscience Conference on Antimicrobial Agents and Chemotherapy 1995 Sep 17; San Francisco, California, 64

  64. Cars O. Efficacy of beta-lactam antibiotics: integration of pharmacokinetics and pharmacodynamics. Diagn Microbiol Infect Dis 1997; 27: 29–33

    Article  PubMed  CAS  Google Scholar 

  65. Craig WA. Choosing an antibiotic on the basis of pharmacodynamics. Ear Nose Throat Journal 1998; 77 Suppl.: 7–12

    CAS  Google Scholar 

  66. Cazzola M, Matera MG, Donner CF. Pharmacokinetics and pharmacodynamics of newer oral cephalosporins: implications for treatment of community-acquired lower respiratory tract infections. Clin Drug Invest 1998; 16: 335–46

    Article  CAS  Google Scholar 

  67. Drusano GL, Goldstein FW. Relevance of the Alexander Project: pharmacodynamic considerations. J Antimicrob Chemother 1996; 38 Suppl. A: 141–54

    Article  PubMed  CAS  Google Scholar 

  68. Owens Jr RC, Tessier P, Nightingale CH, et al. Pharmacodynamics of ceftriaxone and cefixime against community-acquired respiratory tract pathogens. Int J Antimicrob Agents 2001; 17: 483–9

    Article  PubMed  CAS  Google Scholar 

  69. Lutsar I, Ahmed A, Friedl IR, et al. Pharmacodynamics and bactericidal activity of ceftriaxone therapy in experimental cephalosporin-resistant pneumococcal meningitis. Antimicrob Agents Chemother 1997; 41(11): 2414–7

    PubMed  CAS  Google Scholar 

  70. Roche US Pharmaceuticals. Rocephin (ceftriaxone sodium) for injection. 2000 Sep. available from http://www.rocheusa.com/products/rocephin/pi.html (acccessed 15/05/01)

  71. Belliveau PP, Freeman CD, Nicolau DP, et al. Serum bactericidal activity of ceftizoxime and ceftriaxone against pathogens associated with community-acquired and nosocomial pneumonias. Am J Health System Pharm 1996 May 1; 53: 1024–7

    CAS  Google Scholar 

  72. Blandino G, Milazzo I, Musumeci R, et al. Comparative activity of cefodizime and ceftriaxone against respiratory pathogens in an in vitro pharmacodynamic model simulating concentration-time curves. J Chemother 2000; 12: 503–8

    PubMed  CAS  Google Scholar 

  73. Spangler SK, Lin G, Jacobs MR, et al. Postantibiotic effect of sanfetrinem compared with those of six other agents against 12 penicillin-susceptible and -resistant pneumococci. Antimicrob Agents Chemother 1997 Oct; 41: 2173–6

    PubMed  CAS  Google Scholar 

  74. Odenholt I, Lowdin E, Cars O. In vitro pharmacodynamic studies of L-749,345 in comparison with imipenem and ceftriaxone against Gram-positive and Gram-negative bacteria. Antimicrob Agents Chemother 1998 Sep; 42: 2365–70

    PubMed  CAS  Google Scholar 

  75. Patel IH, Kaplan SA. Pharmacokinetic profile of ceftriaxone in man. Am J Med 1984; 19: 17–25

    Google Scholar 

  76. Yuk JH, Nightingale CH, Quintiliani R. Clinical pharmacokinetics of ceftriaxone. Clin Pharmacokinet 1989 Oct; 17: 223–35

    Article  PubMed  CAS  Google Scholar 

  77. Borner K, Lode H, Hampel B, et al. Comparative pharmacokinetics of ceftriaxone after subcutaneous and intravenous administration. Chemotherapy 1985; 31: 237–45

    Article  PubMed  CAS  Google Scholar 

  78. Stoeckel K, Koup JR. Pharmacokinetics of ceftriaxone in patients with renal and liver insufficiency and correlations with a physiologic nonlinear protein binding model. Am J Med 1984 Oct 19; 77: 26–32

    PubMed  CAS  Google Scholar 

  79. Hayton W, Stoeckel K. Biliary excretion of ceftriaxone [letter]. Eur J Clin Pharmacol 1986; 31(1): 123–4

    Article  PubMed  CAS  Google Scholar 

  80. Scaglione F, Raichi M, Fraschini F. Serum protein binding and extravascular diffusion of methoxyimino cephalosporins. Time courses of free and total concentrations of cefotaxime and ceftriaxone in serum and pleural exudate. J Antimicrob Chemother 1990 Sep; 26 Suppl. A: 1–10

    Article  PubMed  Google Scholar 

  81. Adam D, Naber KG. Concentrations of ceftriaxone in prostate adenoma tissue. Chemotherapy 1984; 30: 1–6

    Article  PubMed  CAS  Google Scholar 

  82. De Grandi P, Bauen J-F. Pharmacokinetics of ceftriaxone in serum and gynecological tissues. Chemotherapy 1986; 32: 473–7

    Article  PubMed  Google Scholar 

  83. Joynt GM, Lipman J, Gomersall CD, et al. The pharmacokinetics of once-daily dosing of ceftriaxone in critically ill patients. J Antimicrob Chemother 2001; 47: 421–9

    Article  PubMed  CAS  Google Scholar 

  84. Heinemeyer G, Link J, Weber W, et al. Clearance of ceftriaxone in critical care patients with acute renal failure. Intensive Care Med 1990; 16(7): 448–53

    Article  PubMed  CAS  Google Scholar 

  85. Holazo AA, Patel IH, Weinfeld RE, et al. Ceftriaxone pharmacokinetics following multiple intramuscular dosing. Eur J Clin Pharmacol 1986; 30: 109–12

    Article  PubMed  CAS  Google Scholar 

  86. Lebel M, Grégoire S, Caron M, et al. Difference in blister fluid penetration after single and multiple doses of ceftriaxone. Antimicrob Agents Chemother 1985 Jul; 28(1): 123–7

    Article  PubMed  CAS  Google Scholar 

  87. Fraschini F, Braga PC, Scarpazza G, et al. Human pharmacokinetics and distribution in various tissues of ceftriaxone. Chemotherapy 1986; 32: 192–9

    Article  PubMed  CAS  Google Scholar 

  88. McNamara PJ, Stoeckel K, Ziegler WH. Pharmacokinetics of ceftriaxone following intravenous administration of a 3 g dose. Eur J Clin Pharmacol 1982; 22(1): 71–5

    Article  PubMed  CAS  Google Scholar 

  89. Scully BE, Fu KP, Neu HC. Pharmacokinetics of ceftriaxone after intravenous infusion and intramuscular injection. Am J Med 1984; 77: 112–6

    PubMed  CAS  Google Scholar 

  90. Stoeckel K. Pharmacokinetics of Rocephin, a highly active new cephalosporin with an exceptionally long biological half-life. Chemotherapy 1981; 27 Suppl. 1: 42–6

    Article  PubMed  CAS  Google Scholar 

  91. Stoeckel K, McNamara PJ, Brandt R, et al. Effects of concentration-dependent plasma protein binding on ceftriaxone kinetics. Clin Pharmacol Ther 1981; 29(5): 650–7

    Article  PubMed  CAS  Google Scholar 

  92. Schaad UB, Hayton WL, Stoeckel K. Single dose ceftriaxone kinetics in the newborn. Clin Pharmacol Ther 1985; 37: 522–8

    Article  PubMed  CAS  Google Scholar 

  93. Fiset C, Vallée F, LeBel M, et al. Protein binding of ceftriaxone: comparison of three techniques of determination and the effect of 2-hydroxybenzoylglycine, a drug-binding inhibitor in uremia. Ther Drug Monit 1986; 8: 483–9

    Article  PubMed  CAS  Google Scholar 

  94. Le Van Thoi J, Koumare B, Lhoste F, et al. Human pharmacology of ceftriaxone [in French]. Pathol Biol 1982; 30(6): 345–7

    Google Scholar 

  95. Martin C, Cottin A, Francois-Godfroy N, et al. Concentrations of prophylactic ceftriaxone in abdominal tissues during pancreatic surgery. J Antimicrob Chemother 1997 Sep; 40: 445–8

    Article  PubMed  CAS  Google Scholar 

  96. Hary L, Andrejak M, Leleu S, et al. The pharmacokinetics of ceftriaxone and cefotaxime in cirrhotic patients with ascites. Eur J Clin Pharmacol 1989; 36(6): 613–6

    Article  PubMed  CAS  Google Scholar 

  97. Ducroix JP, Guerlin M, Hary L, et al. Clinical study and pharmacokinetics of ceftriaxone in the treatment of 14 ascitic fluid infections [in French]. Pathol Biol 1988 Oct; 36(8): 1007–10

    PubMed  CAS  Google Scholar 

  98. Orda R, Berger S A, Levy Y, et al. Penetration of ceftriaxone and cefoperazone into bile and gallbladder tissue in patients with acute cholecystitis. Dig Dis Sci 1992 Nov; 37: 1691–3

    Article  PubMed  CAS  Google Scholar 

  99. Berger SA, Levy Y, Halevy A, et al. Penetration of cefazolin, ceftriaxone, cefoperazone, and ceftazidime into human gallbladder tissue and bile. World J Surg 1988; 12: 641–4

    Article  PubMed  CAS  Google Scholar 

  100. Brogard JM, Blickle JF, Jehl F, et al. High biliary elimination of ceftriaxone in man. Int J Clin Pharmacol Ther Toxicol 1988; 26(4): 167–72

    PubMed  CAS  Google Scholar 

  101. Scaglione F, de Martini G, Peretto L, et al. Pharmacokinetic study of cefodizime and ceftriaxone in sera and bones of patients undergoing hip arthroplasty. Antimicrob Agents Chemother 1997; 41(10): 2292–4

    PubMed  CAS  Google Scholar 

  102. Lovering AM, Walsh TR, Bannister GC, et al. The penetration of ceftriaxone and cefamandole into bone, fat and haematoma and relevance of serum protein binding to their penetration into bone. J Antimicrob Chemother 2001; 47: 483–6

    Article  PubMed  CAS  Google Scholar 

  103. Lucht F, Dorche G, Aubert G, et al. The penetration of ceftriaxone into human brain tissue. J Antimicrob Chemother 1990 Jul; 26: 81–6

    Article  PubMed  CAS  Google Scholar 

  104. Kafetzis DA, Brater DC, Fanourgakis JE, et al. Ceftriaxone distribution between maternal blood and fetal blood and tissues at parturition and blood and milk postpartum. Antimicrob Agents Chemother 1983; 23(6): 870–3

    Article  PubMed  CAS  Google Scholar 

  105. Lang R, Saba K, Folman Y, et al. Penetration of ceftriaxone into the intervertebral disc. J Bone Joint Surg Am 1994 May; 76: 689–91

    PubMed  CAS  Google Scholar 

  106. Gaillard J-L, Abadie V, Cheron G, et al. Concentrations of ceftriaxone in cerebrospinal fluid of children with meningitis receiving dexamethasone therapy. Antimicrob Agents Chemother 1994 May; 38: 1209–10

    Article  PubMed  CAS  Google Scholar 

  107. Bradley JS, Farhat C, Stamboulian D, et al. Ceftriaxone therapy of bacterial meningitis: cerebrospinal fluid concentrations and bactericidal activity after intramuscular injection in children treated with dexamethasone. Pediatr Infect Dis J 1994 Aug; 13: 724–8

    Article  PubMed  CAS  Google Scholar 

  108. Nau R, Prange HW, Muth P, et al. Passage of cefotaxime and ceftriaxone into cerebrospinal fluid of patients with uninflamed meninges. Antimicrob Agents Chemother 1993 Jul; 37: 1518–24

    Article  PubMed  CAS  Google Scholar 

  109. Geny F, Costa P, Bressolle F, et al. Ceftriaxone pharmacokinetics in elderly subjects and penetration into epididymis. Biopharm Drug Dispos 1993 Mar; 14: 161–9

    Article  PubMed  CAS  Google Scholar 

  110. Steib A, Jacoberger B, Von Bandel M, et al. Concentrations in plasma and tissue penetration of ceftriaxone and ornidazole during liver transplantation. Antimicrob Agents Chemother 1993 Sep; 37: 1873–6

    Article  PubMed  CAS  Google Scholar 

  111. Lang R, Shalit I, Segal J, et al. Maternal and fetal serum and tissue levels of ceftriaxone following preoperative prophylaxis in emergency cesarean section. Chemotherapy Basel 1993 Mar–Apr; 39: 77–81

    Article  CAS  Google Scholar 

  112. Papaioannou N, Kalivas L, Kalavritinos J, et al. Tissue concentrations of third-generation cephalosporins (ceftazidime and ceftriaxone) in lower extremity tissues using a tourniquet. Arch Orthop Trauma Surg 1994 Apr; 113(3): 167–9

    Article  PubMed  CAS  Google Scholar 

  113. Gudnason T, Gudbrandsson F, Barsanti F, et al. Penetration of ceftriaxone into the middle ear fluid of children. Pediatr Infect Dis J 1998; 17(3): 258–60

    Article  PubMed  CAS  Google Scholar 

  114. Neidhart P, Velebit V, Gunning K, et al. A comparative study of cefamandole and ceftriaxone as prophylaxis in cardiac surgery. Infection 1990; 18(2): 101–4

    Article  PubMed  CAS  Google Scholar 

  115. Gerstner GJ, Kronich W, Adam D. Interstitial fluid concentrations of ceftriaxone (1 g i.v.) in the subperitoneal space after hysterectomy. Chemotherapy Basel 1990 Aug; 36: 245–50

    Article  CAS  Google Scholar 

  116. Berger SA, Dan M, Serour F, et al. Penetration of third-generation cephalosporins into human peritoneal tissue. Chemotherapy 1989; 35: 326–9

    Article  PubMed  CAS  Google Scholar 

  117. Goonetilleke AKE, Dev D, Aziz I, et al. A comparative analysis of pharmacokinetics of ceftriaxone in serum and pleural fluid in humans: a study of once daily administration by intramuscular and intravenous routes. J Antimicrob Chemother 1996 Dec; 38: 969–76

    Article  PubMed  CAS  Google Scholar 

  118. Kimura M, Matsushima T, Nakamura J, et al. Comparative study of penetration of lomefloxacin and ceftriaxone into transudative and exudative pleural effusion. Antimicrob Agents Chemother 1992 Dec; 36: 2774–7

    Article  PubMed  CAS  Google Scholar 

  119. Martin C, Ragni J, Lokiec F, et al. Pharmacokinetics and tissue penetration of a single dose of ceftriaxone (1,000 milligrams intravenously) for antibiotic prophylaxis in thoracic surgery. Antimicrob Agents Chemother 1992 Dec; 36: 2804–7

    Article  PubMed  CAS  Google Scholar 

  120. Sharir M, Triester G, Kneer J, et al. The intravitreal penetration of ceftriaxone in man following systemic administration. Invest Ophthalmol Vis Sci 1989 Oct; 30: 2179–83

    PubMed  CAS  Google Scholar 

  121. Martin E, Koup JR, Paravicini U, et al. Pharmacokinetics of ceftriaxone in neonates and infants with meningitis. J Pediatr 1984 Sep; 105(3): 475–81

    Article  PubMed  Google Scholar 

  122. Martin E. Once-daily administration of ceftriaxone in the treatment of meningitis and other serious infection in children. European Journal of Clinical Microbiology 1983 Oct; 2(5): 509–15

    Article  PubMed  CAS  Google Scholar 

  123. Balant L, Dayer P, Auckenthaler R. Clinical pharmacokinetics of the third generation cephalosporins. Clin Pharmacokinet 1985; 10: 101–43

    Article  PubMed  CAS  Google Scholar 

  124. Pickup ME, Bird HA, Lowe JR, et al. A pharmacokinetic and tolerance study of Ro13-9904, a new cephalosporin antibiotic. Br J Clin Pharmacol 1981; 12: 111–5

    Article  PubMed  CAS  Google Scholar 

  125. Marshall WF, Blair JE. The cephalosporins. Mayo Clin Proc 1999 Feb; 74: 187–95

    Article  PubMed  CAS  Google Scholar 

  126. Koup JR, Keller E, Neumann H, et al. Ceftriaxone pharmacokinetics during peritoneal dialysis. Eur J Clin Pharmacol 1986; 30: 303–7

    Article  PubMed  CAS  Google Scholar 

  127. Stoeckel K, McNamara PJ, Hoppe-Seyler G, et al. Single-dose ceftriaxone kinetics in functionally anephric patients. Clin Pharmacol Ther 1983 May; 33(5): 633–41

    Article  PubMed  CAS  Google Scholar 

  128. Ti T-Y, Fortin L, Kreeft JH, et al. Kinetic disposition of intravenous ceftriaxone in normal subjects and patients with renal failure on hemodialysis or peritoneal dialysis. Antimicrob Agents Chemother 1984 Jan; 25(1): 83–7

    Article  PubMed  CAS  Google Scholar 

  129. Wise R, Wright N. The pharmacokinetics of cefotaxime and ceftriaxone in renal and hepatic dysfunction. Infection 1985; 13 Suppl. 1: S145–50

    Article  PubMed  Google Scholar 

  130. Favre H, Probst P. Pharmacokinetics of ceftriaxone after intravenous administration to CAPD-patients with and without peritonitis. Chemioterapia 1987 Jun; 62 Suppl. 2: 273–4

    Google Scholar 

  131. Hendrickson JR, North DS. Pharmacoeconomic benefit of antibiotic step-down therapy: converting patients from intravenous ceftriaxone to oral cefpodoxime proxetil. Ann Pharmacother 1995 Jun; 29: 561–5

    PubMed  CAS  Google Scholar 

  132. Cammarata SK, Bermudez M, Golin V, et al. Comparison of linezolid versus ceftriaxone/cefpodoxime in the treatment of hospitalized community-acquired pneumonia [abstract]. 9th International Congress on Infectious Diseases 2000 April 10–13; Buenos Aires, Argentina; 182

  133. Cammarata SK, San Pedro GS, Timm JA, et al. Comparison of linezolid versus ceftriaxone/cefpodoxime in the treatment of hospitalized patients with community-acquired pneumonia [abstract]. Clin Microbiol Infect 2000; 6 Suppl. 1: 136

    Google Scholar 

  134. Bittner MJ, Toney JF, Eleftheriou PD, et al. Randomized, double-blind comparison of oral cefpodoxime and parenteral ceftriaxone in hospitalized adults with community-acquired pneumonia. J Clin Outcomes Manage 1999 Mar; 6: 38–45

    Google Scholar 

  135. Nathwani D, Tillotson G, Davey P. Sequential antimicrobial therapy — the role of quinolones. J Antimicrob Chemother 1997; 39: 441–6

    Article  PubMed  CAS  Google Scholar 

  136. Marhoum EFK, Noun M, Chakib A, et al. Ceftriaxone versus penicillin G in the short-term treatment of meningococcal meningitis in adults. Eur J Clin Microbiol Infect Dis 1993 Oct; 12: 766–8

    Article  Google Scholar 

  137. Saez-Llorens X, Feris JM, Klugman KP, et al. Use of a quinolone in children with bacterial meningitis: a comparative study of trovafloxacin and ceftriaxone +/- vancomycin [abstract]. 40th Interscience Conference on Antimicrobial Agents and Chemotherapy 2000 Sep 17; Toronto, Canada, 479

  138. Scholz H, Hofmann T, Noack R, et al. Prospective comparison of ceftriaxone and cefotaxime for the short-term treatment of bacterial meningitis in children. Chemotherapy 1998 Mar–Apr; 44: 142–7

    Article  PubMed  CAS  Google Scholar 

  139. Schmutzhard E, Williams KJ, Vukmirovits G, et al. A randomised comparison of meropenem with cefotaxime or ceftriaxone for the treatment of bacterial meningitis in adults. Meropenem Meningitis Study Group. J Antimicrob Chemother 1995 Jul; 36 Suppl. A: 85–97

    Article  PubMed  CAS  Google Scholar 

  140. Martin E, Hohl P, Guggi T, et al. Short course single daily ceftriaxone monotherapy for acute bacterial meningitis in children: results of a Swiss multicenter study. Part I: clinical results. Infection 1990 Mar–Apr; 18: 70–7

    Article  PubMed  CAS  Google Scholar 

  141. Peltola H, Antilla M, Renkonen O-V, et al. Randomised comparison of chloramphenicol, ampicillin, cefotaxime and ceftriaxone for childhood bacterial meningitis. Lancet 1989 Jun 10: 1281–6

    Article  Google Scholar 

  142. Schaad UB, Suter S, Gianella-Borradori A, et al. A comparison of ceftriaxone and cefuroxime for the treatment of bacterial meningitis in children. N Engl J Med 1990 Jan 18; 322: 141–7

    Article  PubMed  CAS  Google Scholar 

  143. Baraff LJ, Lee SI, Schriger DL. Outcomes of bacterial meningitis in children: a meta-analysis. Pediatr Infect Dis J 1993; 12(5): 389–94

    Article  PubMed  CAS  Google Scholar 

  144. Lebel MH, Hoyt MJ, McCracken Jr GH. Comparative efficacy of ceftriaxone and cefuroxime for treatment of bacterial meningitis. J Pediatr 1989; 114(6): 1049–54

    Article  PubMed  CAS  Google Scholar 

  145. Dowell SF, Butler JC, Giebink GS, et al. Acute otitis media: management and surveillance in an era of pneumococcal resistance — a report from the Drug-resistant Streptococcus pneumoniae Therapeutic Working Group. Pediatr Infect Dis J 1999; 18(1): 1–9

    PubMed  CAS  Google Scholar 

  146. Block SL. Management of acute otitis media in the 1990s: a decade of resistant pneumococcus. Paediatr Drugs 1999 Jan–Mar; 1: 31–50

    Article  PubMed  CAS  Google Scholar 

  147. Leibovitz E, Raiz S, Piglansky L, et al. Resistance pattern of middle ear fluid isolates in acute otitis media recently treated with antibiotics. Pediatr Infect Dis J 1998; 17: 463–9

    Article  PubMed  CAS  Google Scholar 

  148. McCracken Jr GH. Prescribing antimicrobial agents for treatment of acute otitis media. Pediatr Infect Dis J 1999 Dec; 18: 1141–6

    Article  PubMed  Google Scholar 

  149. Klein JO. Review of consensus reports on management of acute otitis media. Pediatr Infect Dis J 1999; 18: 1152–5

    Article  PubMed  CAS  Google Scholar 

  150. Barnett ED, Teele DW, Klein JO, et al. Comparison of ceftriaxone and trimethoprim-sulfamethoxazole for acute otitis media. Greater Boston Otitis Media Study Group. Pediatrics 1997 Jan; 99: 23–8

    Article  PubMed  CAS  Google Scholar 

  151. Green SM, Rothrock SG. Single-dose intramuscular ceftriaxone for acute otitis media in children. Pediatrics 1993 Jan; 91: 23–30

    PubMed  CAS  Google Scholar 

  152. Varsano I, Volovitz B, Horev Z, et al. Intramuscular ceftriaxone compared with oral amoxicillin-clavulanate for treatment of acute otitis media in children. Eur J Pediatr 1997 Nov; 156: 858–63

    Article  PubMed  CAS  Google Scholar 

  153. Cohen R, Navel M, Grunberg J, et al. One dose ceftriaxone vs ten days of amoxicillin/clavulanate therapy for acute otitis media: clinical efficacy and change in nasopharyngeal flora. Pediatr Infect Dis J 1999 May; 18: 403–9

    Article  PubMed  CAS  Google Scholar 

  154. Chamberlain JM, Boenning DA, Waisman Y, et al. Single-dose ceftriaxone versus 10 days of cefaclor for otitis media. Clin Pediatr Phila 1994 Nov; 33: 642–6

    Article  PubMed  CAS  Google Scholar 

  155. Al Ghamdi YS, El Kafrawi M, Aglan YI, et al. Efficacy of single-dose intramuscular ceftriaxone for treatment of acute otitis media in children. Saudi Med J 1999; 20(1): 41–5

    Google Scholar 

  156. Roche Laboratories. Rocephin. In: Murray L, editor. Physicians’ Desk Reference. 55th ed. Montvale: Medical Economics company, 2001. 2765–2768

    Google Scholar 

  157. Bauchner H, Adams W, Barnett E, et al. Therapy for acute otitis media: preference of parents for oral or parenteral antibiotic [see comments]. Arch Pediatr Adolesc Med 1996 Apr; 150: 396–9

    Article  PubMed  CAS  Google Scholar 

  158. Leibovitz E, Piglansky L, Raiz S, et al. Bacteriologic and clinical efficacy of one day vs. three day intramuscular ceftriaxone for treatment of nonresponsive acute otitis media in children. Pediatr Infect Dis J 2000; 19(11): 1040–5

    Article  PubMed  CAS  Google Scholar 

  159. Leibovitz E, Piglansky L, Raiz S, et al. Bacteriologic efficacy of a three-day intramuscular ceftriaxone regimen in non-responsive acute otitis media. Pediatr Infect Dis J 1998 Dec; 17: 1126–31

    Article  PubMed  CAS  Google Scholar 

  160. Gehanno P, Nguyen L, Barry B, et al. Eradication by ceftriaxone of Streptococcus pneumoniae isolates with increased resistance to penicillin in cases of acute otitis media. Antimicrob Agents Chemother 1999 Jan; 43(1): 16–20

    PubMed  CAS  Google Scholar 

  161. Huchon G, Woodhead M. Management of adult community-acquired lower respiratory tract infections. European Study on Community Acquired Pneumonia (EOSCAP) Committee. Eur Resp Rev 1998; 8(61): 391–426

    Google Scholar 

  162. Dowell ME, Mayer H, Anderson A, et al. A randomized, double-blind, multicenter, comparative study of gatifloxacin 400 mg IV and PO versus ceftriaxone plus or minus erythromycin in treatment of community-acquired pneumonia requiring hospitalization [abstract]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy 1999; San Francisco, California; 702

  163. Johnson RH, Levine S, Traub SL, et al. Sequential intravenous/oral ciprofloxacin compared with parenteral ceftriaxone in the treatment of hospitalized patients with community-acquired pneumonia. Infect Dis Clin Pract 1996 May; 5: 265–72

    Google Scholar 

  164. Williams HD, Daniel R, Trovafloxacin Study Group. Trovafloxacin vs ceftriaxone/cepodoxime ± erythromycin in hospitalised community-acquired pneumonia. Drugs 1999; 58 Suppl. 2: 318–9

    Article  Google Scholar 

  165. De Palma M, Rocchi D, Canepa G, et al. Single daily dose of cefodizime in patients with community-acquired pneumonia: an open-label, controlled, randomized study. Clin Ther 1995 May–Jun; 17: 413–24

    Article  PubMed  Google Scholar 

  166. Roson B, Carratala J, Tubau F, et al. Randomized trial of amoxicillin-clavulanate vs ceftriaxone for the treatment of pneumococcal pneumonia in hospitalized patients [abstract]. 38th Interscience Conference on Antimicrobial Agents and Chemotherapy 1998 Sep 24; San Diego, California, 580

  167. Zervos M, Nelson M. Cefepime versus ceftriaxone for empiric treatment of hospitalized patients with community-acquired pneumonia. Cefepime Study Group [see comments]. Antimicrob Agents Chemother 1998 Apr; 42: 729–33

    Article  PubMed  CAS  Google Scholar 

  168. Norrby SR, Petermann W, Willcox PA, et al. A comparative study of levofloxacin and ceftriaxone in the treatment of hospitalized patients with pneumonia. Scand J Infect Dis 1998; 30(4): 397–404

    Article  PubMed  CAS  Google Scholar 

  169. File Jr TM, 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 Sep; 41: 1965–72

    PubMed  CAS  Google Scholar 

  170. Amir J, Harel L, Eidlitz-Markus T, et al. Comparative evaluation of cefixime versus amoxicillin-clavulanate following ceftriaxone therapy of pneumonia. Clin Pediatr 1996 Dec; 35: 629–33

    Article  CAS  Google Scholar 

  171. Leibovitz E, Tabachnik E, Fliedel O, et al. Once-daily intramuscular ceftriaxone in the outpatient treatment of severe community-acquired pneumonia in children [published erratum appears in Clin Pediatr (Phila) 1991 May;30(5):326]. Clin Pediatr Phila 1990 Nov; 29: 634–9

    Article  PubMed  CAS  Google Scholar 

  172. París MM, Ramilo O, McCracken Jr GH. Management of meningitis caused by penicillin-resistant Streptococcus pneumoniae. Antimicrob Agents Chemother 1995 Oct; 39(10): 2171–5

    Article  PubMed  Google Scholar 

  173. Kleiman MB, Weinberg GA, Reynolds JK, et al. Meningitis with beta-lactam-resistant Streptococcus pneumoniae: the need for early repeat lumbar puncture. Pediatr Infect Dis J 1993; 12: 782–3

    Article  PubMed  CAS  Google Scholar 

  174. Bradley JS, Connor JD. Ceftriaxone failure in meningitis caused by Streptococcus pneumoniae with reduced susceptibility to beta-lactam antibiotics. Pediatr Infect Dis J 1991 Nov; 10: 871–3

    Article  PubMed  CAS  Google Scholar 

  175. Friedland IR, Shelton S, Paris M, et al. Dilemmas in diagnosis and management of cephalosporin-resistant Streptococcus pneumoniae meningitis. Pediatr Infect Dis J 1993; 12: 196–200

    Article  PubMed  CAS  Google Scholar 

  176. Lonks JR, Durkin MR, Meyerhoff AN, et al. Meningitis due to ceftriaxone-resistant Streptococcus pneumoniae. N Engl J Med 1995 Mar 30; 332: 893–4

    Article  PubMed  CAS  Google Scholar 

  177. Tan TQ, Schutze GE, Mason Jr EO, et al. Antibiotic therapy and acute outcome of meningitis due to Streptococcus pneumoniae considered intermediately susceptible to broad-spectrum cephalosporins. Antimicrob Agents Chemother 1994 May; 38(5): 918–23

    Article  PubMed  CAS  Google Scholar 

  178. Cabellos C, Viladrich PF, Verdaguer R, et al. Asingle daily dose of ceftriaxone for bacterial meningitis in adults: experience with 84 patients and review of the literature. Clin Infect Dis 1995 May; 20: 1164–8

    Article  PubMed  CAS  Google Scholar 

  179. Friedland IR, Paris M, Ehrett S, et al. Evaluation of antimicrobial regimens for treatment of experimental penicillin- and cephalosporin-resistant pneumococcal meningitis. Antimicrob Agents Chemother 1993 Aug; 37(8): 1630–6

    Article  PubMed  CAS  Google Scholar 

  180. Klugman KP, Friedland IR, Bradley JS. Bactericidal activity against cephalosporin-resistant Streptococcus pneumoniae in cerebrospinal fluid of children with acute bacterial meningitis. Antimicrob Agents Chemother 1995 Sep; 39: 1988–92

    Article  PubMed  CAS  Google Scholar 

  181. Quagliarello VJ, Scheid WM. Treatment of bacterial meningitis. N Engl J Med 1997 Mar 6; 336(10): 708–16

    Article  PubMed  CAS  Google Scholar 

  182. Olivier C, Cohen R, Begué P, et al. Bacteriologic outcome of children with cefotaxime- or ceftriaxone-susceptible and — nonsusceptible Streptococcus pneumoniae meningitis. Pediatr Infect Dis J 2000 Oct; 19(10): 1015–7

    Article  PubMed  CAS  Google Scholar 

  183. Arditi M, Mason Jr EO, Bradley JS, et al. Three-year multicenter surveillance of pneumococcal meningitis in children: clinical characteristics, and outcome related to penicillin susceptibility and dexamethasone use. Pediatrics 1998 Nov; 102: 1087–97

    Article  PubMed  CAS  Google Scholar 

  184. Mogabgab WJ, Lutz FB. Randomized study of cefotaxime versus ceftriaxone for uncomplicated gonorrhea. South Med J 1994 Apr; 87: 461–4

    Article  PubMed  CAS  Google Scholar 

  185. Handsfield HH, McCormack WM, Hook III EW, et al. A comparison of single-dose cefixime with ceftriaxone as treatmentfor uncomplicated gonorrhea. Gonorrhea Treatment Study Group [see comments]. N Engl J Med 1991 Nov 7; 325: 1337–41

    Article  PubMed  CAS  Google Scholar 

  186. Hook III EW, Jones RB, Martin DH, et al. Comparison of ciprofloxacin and ceftriaxone as single-dose therapy for uncomplicated gonorrhea in women. Antimicrob Agents Chemother 1993 Aug; 37: 1670–3

    Article  PubMed  Google Scholar 

  187. Jones RB, Mogabgab WJ, McCormack WM, et al. Randomized comparison of cefotaxime and ceftriaxone in patients with uncomplicated gonorrhea. Clin Ther 1991 Sep–Oct; 13: 550–6

    PubMed  CAS  Google Scholar 

  188. McCormack WM, Mogabgab WJ, Jones RB, et al. Multicenter, comparative study of cefotaxime and ceftriaxone for treatment of uncomplicated gonorrhea. Sex Transm Dis 1993 Sep–Oct; 20: 269–73

    Article  PubMed  CAS  Google Scholar 

  189. Pabst KM, Siegel NA, Smith S, et al. Multicenter, comparative study of enoxacin and ceftriaxone for treatment of uncomplicated gonorrhea. Sex Transm Dis 1989 Jul–Sep; 16: 148–51

    Article  PubMed  CAS  Google Scholar 

  190. Rompalo AM, Colletta L, Caine VA, et al. Efficacy of 250 mg trospectomycin sulfate IM vs 250 mg ceftriaxone IM for treatment of uncomplicated gonorrhea. Sex Transm Dis 1994 Jul–Aug; 21: 213–6

    PubMed  CAS  Google Scholar 

  191. Smith BL, Mogabgab WJ, Dalu ZA, et al. Multicenter trial of fleroxacin versus ceftriaxone in the treatment of uncomplicated gonorrhea. Am J Med 1993 Mar; 94: 81S–4S

    PubMed  CAS  Google Scholar 

  192. Goldstein AMB, Clark JH, Wickler MA. Comparison of single-dose ceftizoxime or ceftriaxone in the treatment of uncomplicated urethral gonorrhea. Sex Transm Dis 1991 Jul–Sep; 18: 180–2

    Article  PubMed  CAS  Google Scholar 

  193. Bryan JP, Hira SK, Brady W, et al. Oral ciprofloxacin versus ceftriaxone for the treatment of urethritis from resistant Neisseria gonorrhoeae in Zambia. Antimicrob Agents Chemother 1990 May; 34(5): 819–22

    Article  PubMed  CAS  Google Scholar 

  194. Baddour LM, Busby L, Shapiro E, et al. Evaluation of treatment with single-dose ampicillin/sulbactam with probenecid or ceftriaxone in patients with uncomplicated gonorrhea. Sex Transm Dis 1992 Nov–Dec; 19: 341–5

    Article  PubMed  CAS  Google Scholar 

  195. Hellmann NS, Nsubuga PS, Baingana-Baingi DJ, et al. Single-dose ampicillin/sulbactam versus ceftriaxone as treatment for uncomplicated gonorrhoea in a Ugandan STD clinic population with a high prevalence of PPNG infection. J Trop Med Hyg 1995 Apr; 98: 95–100

    PubMed  CAS  Google Scholar 

  196. Mroczkowski TF, Millikan LE, Martin DH, et al. Treatment of gonococcal infections with a single 250 mg intramuscular injection of trospectomycin sulphate vs ceftriaxone sodium. Drugs Exp Clin Res 1993; 19: 41–6

    PubMed  CAS  Google Scholar 

  197. Plourde PJ, Tyndall M, Agoki E, et al. Single-dose cefixime versus single-dose ceftriaxone in the treatment of antimicrobial-resistant Neisseria gonorrhoeae infection. J Infect Dis 1992 Oct; 166: 919–22

    Article  PubMed  CAS  Google Scholar 

  198. Portilla I, Lutz B, Montalvo M, et al. Oral cefixime versus intramuscular ceftriaxone in patients with uncomplicated gonococal infections. Sex Transm Dis 1992 Mar–Apr; 19: 94–8

    PubMed  CAS  Google Scholar 

  199. Freedman LD. Reduced dosage of ceftriaxone for uncomplicated gonorrhea in women. J Fam Pract 1990 Aug; 31: 201–2, 205

    PubMed  CAS  Google Scholar 

  200. Centers for Disease Control and Prevention. 1998 guidelines for treatment of sexually transmitted diseases. Morbidity and Mortality Weekly Report 1998 Jan; 47(R-1): 1–118

    Google Scholar 

  201. Arredondo JL, Diaz V, Gaitan H, et al. Oral clindamycin and ciprofloxacin versus intramuscular ceftriaxone and oral doxycycline in the treatment of mild-to-moderate pelvic inflammatory disease in outpatients. Clin Infect Dis 1997 Feb; 24: 170–8

    Article  PubMed  CAS  Google Scholar 

  202. Sanchez-Ramos L, McAlpine KJ, Adair CD, et al. Pyelonephritis in pregnancy: once-a-day ceftriaxone versus multiple doses of cefazolin: a randomized, double-blind trial. Am J Obstet Gynecol 1995 Jan; 172 (Pt 1): 129–33

    Article  PubMed  CAS  Google Scholar 

  203. Wing DA, Hendershott CM, Debuque L, et al. A randomized trial of three antibiotic regimens for the treatment of pyelonephritis in pregnancy. Obstet Gynecol 1998 Aug; 92: 249–53

    Article  PubMed  CAS  Google Scholar 

  204. Wing DA, Hendershott CM, Debuque L, et al. Outpatient treatment of acute pyelonephritis in pregnancy after 24 weeks. Obstet Gynecol 1999 Nov; 94: 683–8

    Article  PubMed  CAS  Google Scholar 

  205. Francois P, Bensman A, Begue P, et al. Assessment of the efficacy and cost efficiency of two strategies in the treatment of acute pyelonephritis in children: Oral cefixime or parenteral ceftriaxone after an initial IV combination therapy [in French]. Med Mal Infect 1997 Jun; 27 Spec. Iss.: 667–73

    Article  Google Scholar 

  206. Millar LK, Wing DA, Paul RH, et al. Outpatient treatment of pyelonephritis in pregnancy: a randomized controlled trial. Obstet Gynecol 1995 Oct; 86 (Pt 1): 560–4

    PubMed  CAS  Google Scholar 

  207. Wilimas JA, Flynn PM, Harris S, et al. A randomized study of outpatient treatment with ceftriaxone for selected febrile children with sickle cell disease. N Engl J Med 1993 Aug 12; 329: 472–6

    Article  PubMed  CAS  Google Scholar 

  208. Williams LL, Wilimas JA, Harris SC, et al. Outpatient therapy with ceftriaxone and oral cefixime for selected febrile children with sickle cell disease. J Pediatr Hematol Oncol 1996 Aug; 18: 257–61

    Article  PubMed  CAS  Google Scholar 

  209. Leibovitz E, Janco J, Piglansky L, et al. Oral ciprofloxacin vs. intramuscular ceftriaxone as empiric treatment of acute invasive diarrhea in children. Pediatr Infect Dis J 2000; 19(11): 1060–7

    Article  PubMed  CAS  Google Scholar 

  210. Girgis NI, Sultan Y, Hammad O, et al. Comparison of the efficacy, safety and cost of cefixime, ceftriaxone and aztreonam in the treatment of multidrug-resistant Salmonella typhi septicemia in children. Pediatr Infect Dis J 1995 Jul; 14: 603–5

    Article  PubMed  CAS  Google Scholar 

  211. Frenck Jr RW, Nakhla I, Sultan Y, et al. Azithromycin versus ceftriaxone for the treatment of uncomplicated typhoid fever in children. Clin Infect Dis 2000 Nov; 31: 1134–8

    Article  PubMed  CAS  Google Scholar 

  212. Kulhanjian J, Dunphy MG, Hamstra S, et al. Randomized comparative study of ampicillin/sulbactam vs. ceftriaxone for treatment of soft tissue and skeletal infections in children. Pediatr Infect Dis J 1989 Sep; 8: 605–10

    Article  PubMed  CAS  Google Scholar 

  213. Van Reempts PJ, Van Overmeire B, Mahieu LM, et al. Clinical experience with ceftriaxone treatmetn in the neonate. Chemotherapy 1995; 41: 316–22

    Article  PubMed  Google Scholar 

  214. Bass JW, Steele RW, Wittler RR, et al. Antimicrobial treatment of occult bacteremia: a multicenter cooperative study. Pediatr Infect Dis J 1993 Jun; 12(6): 466–73

    Article  PubMed  CAS  Google Scholar 

  215. Fleisher GR, Rosenberg N, Vinci R, et al. Intramuscular versus oral antibiotic therapy for the prevention of meningitis and other bacterial sequelae in young, febrile children at risk for occult bacteremia. J Pediatr 1994 Apr; 124(4): 504–12

    Article  PubMed  CAS  Google Scholar 

  216. Simmons BP, Gelfand MS, Grogan J, et al. Cefotaxime twice daily versus ceftriaxone once daily: a randomized controlled study in patients with serious infections. Diagn Microbiol Infect Dis 1995 May–Jun; 22: 155–7

    Article  PubMed  CAS  Google Scholar 

  217. Shah PM, Stille W, German PSG. Cefotaxime versus ceftriaxone for the treatment of nosocomial pneumonia: results of a multicenter study. Diagn Microbiol Infect Dis 1995 May–Jun; 22: 171–2

    Article  PubMed  CAS  Google Scholar 

  218. Bassetti D, Cruciani M, Solbiati M, et al. Comparative efficacy of ceftriaxone versus ceftazidime in the treatment of nosocomial lower respiratory tract infections. Chemotherapy 1991; 37: 371–5

    Article  PubMed  CAS  Google Scholar 

  219. ter Braak EW, de Vries PJ, Bouter KP, et al. Once-daily dosing regimen for aminoglycoside plus β-lactam combination therapy of serious bacterial infections: comparative trial with netilmicin plus ceftriaxone. Am J Med 1990; 89(1): 58–66

    Article  PubMed  Google Scholar 

  220. Rolston KVI, Jones PG, Fainstein V, et al. Single agent therapy for infections in cancer patients: a prospective randomized trial comparing 3 extended-spectrum cephalosporins. Eur J Clin Microbiol Infect Dis 1991; 3: 139–45

    Article  Google Scholar 

  221. Petrilli AS, Melaragno R, Barros KVT, et al. Fever and neutropenia in children with cancer: a therapeutic approach related to the underlying disease. Pediatr Infect Dis J 1993; 12: 916–21

    Article  PubMed  CAS  Google Scholar 

  222. D’Antonio D, Fioritoni G, Iacone A, et al. Randomized comparison of ceftriaxone versus ceftriaxone plus amikacin for the empirical treatment of infections in patients with altered host defences: microbiological and clinical evaluation. Chemotherapy 1992; 38: 420–7

    Article  PubMed  Google Scholar 

  223. Wang L-S, Cheng D-L, Hinthorn DR, et al. Short-course treatment of bacteremia with ceftriaxone monotherapy. J Formos Med Assoc 1990 Feb; 89: 115–20

    PubMed  CAS  Google Scholar 

  224. Thomas PD, Daly S, Misan G, et al. Comparison of the efficacy and adverse effect profile of cefotaxime, 3 g.day, and ceftriaxone, 2 g.day, in the treatment of nosocomial lower respiratory tract infections in ICU patients. Eur Resp Rev 1994 Sep; 4(22): 321–8

    Google Scholar 

  225. Mandell LA, Bergeron MG, Ronald AR, et al. Once-daily therapy with ceftriaxone compared with daily multiple-dose therapy with cefotaxime for serious bacterial infections: a randomized, double-blind study. J Infect Dis 1989 Sep; 160(3): 433–41

    Article  PubMed  CAS  Google Scholar 

  226. Rubinstein E, Lode H, Grassi C. Ceftazidime monotherapy vs. ceftriaxone/tobramycin for serious hospital-acquired gramnegative infections. Antibiotic Study Group [published erratum appears in Clin Infect Dis 1995 Jun;20(6):1582]. Clin Infect Dis 1995 May; 20: 1217–28

    Article  PubMed  CAS  Google Scholar 

  227. American Thoracic Society. Hospital-acquired pneumonia in adults: diagnosis, assessment of severity, initial antimicrobial therapy, and preventative strategies: a consensus statement. Am J Respir Crit Care Med 1996; 153: 1711–25

    Google Scholar 

  228. Mangi RJ, Peccerillo KM, Ryan J, et al. Cefoperazone versus ceftriaxone monotherapy of nosocomial pneumonia. Diagn Microbiol Infect Dis 1992 Jul; 15: 441–7

    Article  PubMed  CAS  Google Scholar 

  229. Dormann AJ, Wigginghaus B, Risius H, et al. A single dose of ceftriaxone administered 30 minutes before percutaneous endoscopic gastrostomy significantly reduces local and systemic infective complications. Am J Gastroenterol 1999 Nov; 94: 3220–4

    Article  PubMed  CAS  Google Scholar 

  230. Hjortrup A, Moesgaard F, Jensen F, et al. Antibiotic prophylaxis in high risk biliary surgery: one dose of ceftriaxone compared with two doses of cefuroxime. Eur J Surg 1991 Jun–Jul; 157: 403–5

    PubMed  CAS  Google Scholar 

  231. Luke M, Iversen J, Sondergaard J, et al. Ceftriaxone vs. ampicillin + metronidazole as prophylaxis against infections after clean-contaminated abdominal surgery. Eur J Surg 1991 Jan; 157: 45–9

    PubMed  CAS  Google Scholar 

  232. Thomas R, Alvino P, Cortino GR, et al. Long-acting versus short-acting cephalosporins for preoperative prophylaxis in breast surgery: a randomized double-blind trial involving 1,766 patients. Chemotherapy 1999; 45: 217–23

    Article  PubMed  CAS  Google Scholar 

  233. Morris WT. Cetriaxone is more effective than gentamicin/metronidazole prophylaxis in reducing wound and urinary tract infections after bowel operations. Results of a controlled, randomized, blind clinical trial. Dis Colon Rectum 1993 Sep; 36: 826–33

    Article  PubMed  CAS  Google Scholar 

  234. Henriques CU, Wilken-Jensen C, Thorsen P, et al. Arandomised controlled trial of prophylaxis of post-abortal infection: ceftriaxone versus placebo [see comments]. Br J Obstet Gynaecol 1994 Jul; 101: 610–4

    Article  PubMed  CAS  Google Scholar 

  235. von-Mandach U, Huch R, Malinverni R, et al. Ceftriaxone (single dose) versus cefoxitin (multiple doses): success and failure of antibiotic prophylaxis in 1052 cesarean sections. J Perinat Med 1993; 21: 385–97

    Article  Google Scholar 

  236. Boxma H, Broekhuizen T, Patka P, et al. Randomised controlled trial of single-dose antibiotic prophylaxis in surgical treatment of closed fractures: the Dutch Trauma Trial. Lancet 1996 Apr 27; 347: 1133–7

    Article  PubMed  CAS  Google Scholar 

  237. Castelao AM, Soto K, Grinyo JM, et al. Prophylaxis of urinary tract infection in renal transplantation: comparison of three different protocols using ceftriaxone-cloxacillin, aztreonam-cloxacillin, or aztreonam-amoxycillin-clavulanic acid. Transplant Proc 1995 Aug; 27: 2277–9

    PubMed  CAS  Google Scholar 

  238. Raz R, Almog D, Elhanan G, et al. The use of ceftriaxone in the prevention of urinary tract infection in patients undergoing transurethral resection of the prostate (TUR-P). Infection 1994 Sep–Oct; 22: 347–9

    Article  PubMed  CAS  Google Scholar 

  239. Lippert H, Gastinger J. Antimicrobial prophylaxis in laproscopic and conventional cholecystectomy:conclusions of a large prospective multicenter quality assurance study in Germany. Chemotherapy 1998; 44: 355–63

    Article  PubMed  CAS  Google Scholar 

  240. Bhuva M, Ganger D, Jensen D. Spontaneous bacterial peritonitis: an update on evaluation, management, and prevention. Am J Med 1994 Aug; 97: 169–75

    Article  PubMed  CAS  Google Scholar 

  241. Gomez-Jimenez J, Ribera E, Gasser I, et al. Randomized trial comparing ceftriaxone with cefonicid for treatment of spontaneous bacterial peritonitis in cirrhotic patients. Antimicrob Agents Chemother 1993 Aug; 37: 1587–92

    Article  PubMed  CAS  Google Scholar 

  242. Javid G, Khan BA, Khan BA, et al. Short-course ceftriaxone therapy in spontaneous bacterial peritonitis. Postgrad Med J 1998 Oct; 74: 592–5

    Article  PubMed  CAS  Google Scholar 

  243. Francioli P, Etienne J, Hoigné R, et al. Treatment of streptococcal endocarditis with a single daily dose of ceftriaxone sodium for 4 weeks: efficacy and outpatient treatment feasibility. JAMA 1992 Jan 8; 267(2): 264–7

    Article  PubMed  CAS  Google Scholar 

  244. Sexton DJ, Tenenbaum MJ, Wilson WR, et al. Ceftriaxone once daily for four weeks compared with ceftriaxone plus gentamicin once daily for two weeks for treatment of endocarditis due to penicillin-susceptible streptococci. Clin Infect Dis 1999 Dec; 27: 1470–4

    Article  Google Scholar 

  245. Fekety FR. Safety of parenteral third-generation cephalosporins. Am J Med 1990; 88 Suppl. 4A: 38S–44S

    Article  PubMed  CAS  Google Scholar 

  246. Roche Products Limited. Rocephin. In: ABPI Compendium of Data Sheets and Summaries of Product Characteristics. 1999–2000: 1357–9

  247. Kim YS, Kestell MF, Lee SP. Gall-bladder sludge: lessons from ceftriaxone. J Gastroenterol Hepatol 1992; 7: 618–21

    Article  PubMed  CAS  Google Scholar 

  248. Shiffman ML, Keith FB, Moore EW. Pathogenesis of ceftriaxone-associated biliary sludge: in vitro studies of calcium-ceftriaxone binding and solubility. Gastroenterology 1990; 99(6): 1772–8

    PubMed  CAS  Google Scholar 

  249. Bonnet JP, Abid L, Dabhar A, et al. Early biliary pseudolithiasis during ceftriaxone therapy for acute pyelonephritis in children: a prospective study in 34 children. Eur J Pediatr Surg 2000; 10: 368–71

    PubMed  CAS  Google Scholar 

  250. Palanduz A, Yalçin I, Tonguç E, et al. Sonographic assessment of ceftriaxone-associated biliary pseudolithiasis in children. J Clin Ultrasound 2000 May; 28(4): 166–8

    Article  PubMed  CAS  Google Scholar 

  251. Schaad UB, Wedgwood-Krucko J, Tschaeppeler H. Reversible ceftriaxone-associated biliary pseudolithiasis in children. Lancet 1988 Dec 17: 1411–3

    Article  Google Scholar 

  252. Stabile A, Ferrara P, Marietti G, et al. Ceftriaxone-associated gallbladder lithiasis in children [letter]. Eur J Pediatr 1995 Jul; 154: 590

    Article  PubMed  CAS  Google Scholar 

  253. Heim-Duthoy KL, Caperton EM, Pollock R, et al. Apparent biliary pseudolithiasis during ceftriaxone therapy. Antimicrob Agents Chemother 1990; 34(6): 1146–9

    Article  PubMed  CAS  Google Scholar 

  254. Pigrau C, Pahissa A, Gropper S, et al. Ceftriaxone-associated biliary pseudolithaisis in adults. Lancet 1989 Jul 15: 165

    Article  Google Scholar 

  255. Biais C, Duperval R. Biliary pseudolithiasis in a child associated with 2 days of ceftriaxone therapy. Pediatr Radiol 1994; 24: 218–9

    Article  Google Scholar 

  256. Robertson FM, Crombleholme TM, Barlow SE, et al. Ceftriaxone choledocholthiasis. Pediatrics 1996; 98: 133

    PubMed  CAS  Google Scholar 

  257. Lopez AJ, O’Keefe P, Morrissey M, et al. Ceftriaxone-induced cholelithiasis. Ann Intern Med 1991; 115: 712–4

    PubMed  CAS  Google Scholar 

  258. Zinberg J, Chernaik R, Coman E, et al. Reversible symptomatic biliary obstruction associated with ceftriaxone pseudolithiasis. Am J Gastroenterol 1991; 86(9): 1251–4

    PubMed  CAS  Google Scholar 

  259. Zimmermann AE, Katona BG, Jodhka JS, et al. Ceftriaxone-induced acute pancreatitis. Ann Pharmacother 1993 Jan; 27: 36–7

    PubMed  CAS  Google Scholar 

  260. De Moor RA, Egberts AC-G, Schröder CH. Ceftriaxone-associated nephrolithiasis and biliary pseudolithiasis. Eur J Pediatr 1999 Dec; 158: 975–7

    Article  PubMed  Google Scholar 

  261. Grasberger H, Otto B, Loeschke K. Ceftriaxone-associated nephrolithiasis [1] [letter]. Ann Pharmacother 2000; 34(9): 1076–7

    Article  PubMed  CAS  Google Scholar 

  262. Scimeca PG, Weinblatt ME, Boxer R. Hemolysis after treatment with ceftriaxone [1]. J Pediatr 1996; 128(1): 163

    PubMed  CAS  Google Scholar 

  263. Bernini JC, Mustafa MM, Sutor LJ, et al. Fatal hemolysis induced by ceftriaxone in a child with sickle cell anemia. J Pediatr 1995; 126: 813–5

    Article  PubMed  CAS  Google Scholar 

  264. Lascari AD, Amyot K. Fatal hemolysis caused by ceftriaxone. J Pediatr 1995; 126: 816–7

    Article  PubMed  CAS  Google Scholar 

  265. Martin E, Fanconi S, Kälin p, et al. Ceftriaxone — bilirubin — albumin interactions in the neonate: an in vivo study. Eur J Pediatr 1993; 152: 530–4

    Article  PubMed  CAS  Google Scholar 

  266. Hayward CJ, Nafziger AN, Kohlhepp SJ, et al. Investigation of bioequivalence and tolerability of intramuscular ceftriaxone injections by using 1 % lidocaine, buffered lidocaine, and sterile water diluents. Antimicrob Agents Chemother 1996 Feb; 40: 485–7

    PubMed  CAS  Google Scholar 

  267. Schichor A, Bernstein B, Weinerman H, et al. Lidocaine as a diluent for ceftriaxone in the treatment of gonorrhea: does it reduce the pain of the injection? Arch Pediatr Adolesc Med 1994 Jan; 148: 72–5

    Article  PubMed  CAS  Google Scholar 

  268. Patel IH, Weinfeld RE, Konikoff J, et al. Pharmacokinetics and tolerance of ceftriaxone in humans after single-dose intramuscular administration in water and lidocaine diluents. Antimicrob Agents Chemother 1982; 21(6): 957–62

    Article  PubMed  CAS  Google Scholar 

  269. Roche Laboratories. Rocephin (ceftriaxone sodium) for injection. In: Physician’s desk reference. 54th ed. 2000: 2652–4

  270. Roche Pharmaceuticals. Rocephin (ceftriaxone sodium) for injection. 2000 Sep

  271. Tauber MG. Management of bacterial meningitis in adults. Curr Opin Crit Care 1998 Oct; 4: 276–81

    Article  Google Scholar 

  272. Friedland IR, McCracken Jr GH. Management of infections caused by antibiotic-resistant Streptococcus pneumoniae. N Engl J Med 1994 Aug 11; 331(6): 377–82

    Article  PubMed  CAS  Google Scholar 

  273. Heffelfinger JD, Dowell SF, Jorgenson JH, et al. Management of community-acquired pneumonia in the era of pneumococcal resistance: a report from the Drug-ResistanT Streptococcus pneumoniae Therapeutic Working Group. Arch Intern Med 2000; 160: 1399–408

    Article  PubMed  CAS  Google Scholar 

  274. Bradley JS. Oral vs. intramuscular antibiotic therapy for acute otitis media: which is best? Pediatr Infect Dis J 1999; 18(12): 1147–51

    Article  PubMed  CAS  Google Scholar 

  275. Williams DN, Rehm SJ, Tice AD, et al. Practice guidelines for community-based parenteral anti-infective therapy. Clin Infect Dis 1997 Oct; 25: 787–801

    Article  PubMed  CAS  Google Scholar 

  276. Craig WA. Antibiotic selection factors and description of a hospital-based outpatient antibiotic therapy program in the USA. Eur J Clin Microbiol Infect Dis 1995; 14(7): 636–42

    Article  PubMed  CAS  Google Scholar 

  277. Tice AD. Experience with a physician-directed, clinic-based program for outpatient parenteral antibiotic therapy in the USA. Eur J Clin Microbiol Infect Dis 1995; 14(7): 655–61

    Article  PubMed  CAS  Google Scholar 

  278. Barman Balfour JA, Lamb HM. Outpatient parenteral antibacterial therapy of serious infections: the role of ceftriaxone. Dis Manage Health Outcomes 1998 Oct; 4(4): 225–36

    Article  Google Scholar 

  279. International Antimicrobial Therapy Cooperative Group of the European Organization for Research and Treatment of Cancer. Efficacy and toxicity of single daily doses of amikacin and ceftriaxone versus multiple daily doses of amikacin and ceftazidime for infection in patients with cancer and granulocytopenia. Ann Intern Med 1993 Oct 1; 119 (7 Pt 1): 584–93

    Google Scholar 

  280. Gold HS, Moellering Jr RC. Antimicrobial-drug resistance. N Engl J Med 1996 Nov 7; 335(19): 1445–53

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lesley J. Scott.

Additional information

Various sections of the manuscript reviewed by: S.L. Block, Physicians To Children & Adolescents, Bardstown, Kentucky, USA; J. Lipman, Intensive Care Facility and Department of Anaesthesiology and Critical Care, University of Queensland, Royal Brisbane Hospital, Brisbane, Queensland, Australia; K.G. Naber, Urologische Klinik, Klinikum St. Elisabeth Straubing, Straubing, Germany; R. Nau, Georg-August-Universitat Göttingen, Göttingen, Germany; F. Scaglione, Department of Pharmacology, Chemotherapy and Toxicology, Faculty of Medicine, University of Milan, Milan, Italy; K. Weiss, Hopital Maisonneuve-Rosemont, University of Montreal, Montreal, Quebec, Canada.

Data Selection

Sources: Medical literature published in any language since 1988 on ceftriaxone, identified using Medline, supplemented by AdisBase (a proprietary database of Adis International). Additional references were identified from the reference lists of published articles. Bibliographical information, including contributory unpublished data, was also requested from the company developing the drug.

Search strategy: Medline search terms were ‘ceftriaxone’ or ‘ceftriaxone sodium’ and (lower respiratory tract infections or pneumonia or bronchitis or AECB or acute otitis media or bacterial otitis media or skin structure infections or urinary tract infections or uncomplicated gonorrhoea or bacterial septicaemia or bone and joint infections or intra-abdominal infections or meningitis-bacterial or surgical prophylaxis or febrile neutropenia or pharmacoeconomics). AdisBase search terms were ‘ceftriaxone’ and (lower respiratory tract infections or pneumonia or bronchitis or AECB or acute otitis media or skin structure infections or urinary tract infections or uncomplicated gonorrhoea or pelvic inflammatory disease or bacterial septicaemia or bone-and-joint-infections or osteomyelitis or infectious arthritis or meningitis or surgical prophylaxis or febrile neutropenia). Searches were last updated 13 Mar 2002.

Selection: Studies in patients with community-acquired or nosocomial infections who received ceftriaxone. Inclusion of studies was based mainly on the methods section of the trials. When available, large, well-controlled, randomised trials with appropriate statistical methodology were preferred. Relevant in vitro and pharmacokinetic data are also included.

Index terms: Community-acquired pneumonia, otitis media, meningitis, sexually transmitted infections, acute pyelonephritis, paediatric infections, nosocomial infections, surgical prophylaxis, antibacterial activity, pharmacokinetics, therapeutic use, peritonitis.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lamb, H.M., Ormrod, D., Scott, L.J. et al. Ceftriaxone. Drugs 62, 1041–1089 (2002). https://doi.org/10.2165/00003495-200262070-00005

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-200262070-00005

Keywords

Navigation