Skip to main content

Cephalosporins General Statement (Monograph)

Drug class: Cephalosporins
VA class: AM100

Introduction

Cephalosporins are semisynthetic β-lactam antibiotics that are structurally and pharmacologically related to penicillins and cephamycins (e.g., cefotetan, cefoxitin). Cephalosporins generally are divided into 5 groups (“generations”) based on their spectra of activity.

Uses for Cephalosporins General Statement

Cephalosporins are used parenterally for the treatment of lower respiratory tract, skin and skin structure, urinary tract, and bone and joint infections caused by susceptible gram-positive or gram-negative bacteria and also are used parenterally for the treatment of meningitis and septicemia/bacteremia caused by susceptible gram-positive or gram-negative bacteria. Cephalosporins also are used parenterally for the treatment of intra-abdominal, biliary tract, and gynecologic infections (including pelvic inflammatory disease) caused by susceptible bacteria. Cefotaxime, cefoxitin, ceftriaxone, and cefuroxime are used parenterally for the treatment of uncomplicated gonorrhea or other gonococcal infections; cefepime, ceftazidime, and ceftriaxone are used for empiric anti-infective therapy in febrile neutropenic patients; and cefazolin, cefotaxime, ceftriaxone, and cefuroxime are used parenterally for perioperative prophylaxis.

Cephalosporins are used orally for the treatment of mild to moderate respiratory tract infections, including acute maxillary sinusitis, acute bacterial exacerbations of chronic bronchitis, secondary infections of acute bronchitis, and community-acquired pneumonia, caused by susceptible bacteria (e.g., Streptococcus pneumoniae, Haemophilus influenzae, H. parainfluenzae, Moraxella catarrhalis);307 311 338 339 341 348 663 664 677 acute bacterial otitis media caused by susceptible bacteria (e.g., S. pneumoniae, H. influenzae, M. catarrhalis);307 311 338 339 341 348 677 and pharyngitis and tonsillitis caused by Streptococcus pyogenes (group A β-hemolytic streptococci).307 311 338 339 341 348 663 664 677 Cefaclor, cefadroxil, cefdinir, cefditoren pivoxil, cefpodoxime proxetil, cefprozil, ceftibuten, cefuroxime axetil, and cephalexin also are used orally for the treatment of mild to moderate skin and skin structure infections caused by susceptible staphylococci or streptococci.307 308 311 338 339 341 348 663 664 In addition, cefaclor, cefadroxil, cefixime, cefpodoxime proxetil, cefuroxime axetil, and cephalexin are used orally for the treatment of mild to moderate urinary tract infections caused by susceptible gram-negative bacteria (e.g., Escherichia coli, Klebsiella, Proteus mirabilis).307 308 341 348 677 Some clinicians suggest that certain oral third generation cephalosporins (cefdinir, cefpodoxime proxetil, ceftibuten) are one of several alternatives that can be used for the outpatient treatment of recurrent urinary tract infections or urinary tract infections acquired in hospitals or nursing homes since these infections are likely to be caused by multidrug-resistant gram-negative bacilli.710 Certain oral cephalosporins (e.g., cefixime, cefpodoxime proxetil, cefuroxime axetil) have been used for the treatment of uncomplicated gonorrhea.292 338 344 348 677 (See Uses: Gonorrhea and Associated Infections.)

Prior to and during cephalosporin therapy, the causative organism should be cultured and in vitro susceptibility tests performed. In serious infections, therapy may be initiated pending results of in vitro tests. In certain serious infections when the causative organism is unknown, concomitant therapy with another anti-infective agent (e.g., an aminoglycoside) may be indicated pending results of susceptibility tests. Use of a cephalosporin does not replace surgical procedures such as incision and drainage when indicated.338

Gram-positive Bacterial Infections

First and second generation cephalosporins are used in the treatment of infections caused by susceptible staphylococci or streptococci.307 308 339 348 Third generation cephalosporins generally are less active than first and second generation cephalosporins against gram-positive aerobic bacteria, especially staphylococci, and usually are not used in the treatment of infections caused by gram-positive bacteria when a penicillin or a first or second generation cephalosporin could be used.453 Some third generation cephalosporins (e.g., cefotaxime, ceftriaxone) are considered drugs of choice for serious bacterial infections, including endocarditis or meningitis, caused by susceptible S. pneumoniae or viridans streptococci. Unlike other cephalosporins, fifth generation cephalosporins (e.g., ceftaroline) have activity against methicillin-resistant S. aureus (MRSA, also known as oxacillin-resistant S. aureus [ORSA]) and are used in the treatment of some MRSA infections.704 705 711 712 713 715

Gram-negative Bacterial Infections

Use of first generation cephalosporins (cefazolin) in the treatment of gram-negative bacterial infections generally is limited to infections caused by susceptible E. coli, H. influenzae, Klebsiella, or P. mirabilis. Second and third generation cephalosporins are used in the treatment of infections caused by these organisms as well as infections caused by susceptible Enterobacter, Morganella morganii (formerly Proteus morganii), Neisseria, Providencia rettgeri (formerly P. rettgeri), or P. vulgaris; cefotaxime, ceftazidime, and ceftriaxone also are used in the treatment of infections caused by susceptible Serratia. Certain parenteral third generation cephalosporins (i.e., cefepime, cefotaxime, ceftazidime, ceftriaxone) may be drugs of choice for the treatment of infections caused by susceptible Enterobacteriaceae, including susceptible E. coli, Klebsiella pneumoniae, P. rettgeri, M. morganii, P. vulgaris, or P. stuartii and are alternatives for the treatment of infections caused by susceptible Serratia; an aminoglycoside usually is used concomitantly in severe infections.710 Ceftazidime and cefepime are considered alternatives for the treatment of infections caused by susceptible Pseudomonas aeruginosa; an aminoglycoside may be used concomitantly.710 Ceftazidime is more active in vitro on a weight basis against Ps. aeruginosa than most other currently available cephalosporins and is active against some strains resistant to many other cephalosporins, aminoglycosides, and extended-spectrum penicillins. Ceftaroline, a fifth generation cephalosporin, generally has activity against gram-negative aerobes that is similar to that reported with third generation cephalosporins.704

Endocarditis

Treatment

Ceftriaxone is used for the treatment of native valve or prosthetic valve endocarditis caused by viridans streptococci (e.g., S. oralis, S. milleri group, S. mitis, S. mutans, S. salivarius, S. sanguis, Gamella morbillorum) or S. bovis (nonenterococcal group D streptococcus) [off-label].450 685 The drug also is used for the treatment of native valve or prosthetic valve endocarditis caused by slow-growing fastidious gram-negative bacilli termed the HACEK group [off-label] (i.e., Haemophilus parainfluenzae, H. aphrophilus, H. paraphrophilus, H. influenzae, Actinobacillus actinomycetemcomitans, Cardiobacterium hominis, Eikenella corrodens, Kingella kingae, K. denitrificans).450 In addition, ceftriaxone has been used for the treatment of native or prosthetic valve endocarditis caused by E. faecalis resistant to penicillin, aminoglycosides, and vancomycin.450

IV cefazolin is used as an alternative to nafcillin or oxacillin for the treatment of staphylococcal endocarditis, including infections caused by coagulase-positive strains (S. aureus) or coagulase-negative strains (e.g., S. epidermidis [off-label], S. lugdunensis [off-label]) in penicillin-allergic patients (nonanaphylactoid type only).450 685 Cefazolin also is used as an alternative to penicillin G sodium for the treatment of endocarditis caused by susceptible Streptococcus pyogenes (group A β-hemolytic streptococci)310 450 or S. pneumoniae [off-label].450

For empiric treatment of culture-negative endocarditis in prosthetic valve recipients with early onset endocarditis (within 1 year after prosthetic valve placement), a multiple-drug regimen that includes vancomycin, gentamicin, cefepime, and rifampin is recommended.450 )

Prevention

Certain cephalosporins (cefazolin, ceftriaxone, cephalexin, cefadroxil) are used as alternatives to amoxicillin or ampicillin for prevention of α-hemolytic (viridans group) streptococcal endocarditis in penicillin-allergic adults and children undergoing certain dental or upper respiratory tract procedures who have underlying cardiac conditions that put them at the highest risk of adverse outcome from endocarditis.451 These cephalosporins should not be used for such prophylaxis in patients with a history of immediate-type penicillin hypersensitivity (e.g., urticaria, angioedema, anaphylaxis).451

The cardiac conditions identified by the American Heart Association (AHA) as those associated with the highest risk and for which endocarditis prophylaxis is recommended are prosthetic cardiac valves, previous infective endocarditis, congenital heart disease (i.e., unrepaired cyanotic congenital heart disease including palliative shunts and conduits; a completely repaired congenital heart defect where a prosthetic material or device was placed by surgery or catheter intervention within the last 6 months; repaired congenital heart disease with residual defects at the site or adjacent to the site of a prosthetic patch or prosthetic device which inhibits endothelialization), and cardiac valvulopathy after cardiac transplantation.451 AHA states that endocarditis prophylaxis is recommended for such patients when they undergo dental procedures that involve manipulation of gingival tissue or the periapical region of teeth or perforation of the oral mucosa or undergo invasive procedures of the respiratory tract that involve incision or biopsy of the respiratory mucosa (e.g., tonsillectomy, adenoidectomy).451 Although endocarditis prophylaxis also may be indicated for such patients when they undergo surgical procedures that involve infected skin, skin structure, or musculoskeletal tissue, prophylaxis solely to prevent infective endocarditis is not recommended for GI or genitourinary tract procedures.451

When selecting anti-infectives for prophylaxis of bacterial endocarditis, the current recommendations published by AHA should be consulted.451

GI Infections

Some parenteral third generation cephalosporins are used in the treatment of GI infections caused by Salmonella292 503 or Shigella292 570 673 674 or the treatment of other uncommon infectious diarrheal illnesses, including infections caused by Vibrio503 582 and Yersinia.503

Salmonella Gastroenteritis

Anti-infective therapy generally is not indicated in otherwise healthy individuals with uncomplicated (noninvasive) gastroenteritis caused by non-typhi Salmonella (e.g., S. enteritidis, S. typhimurium) since these infections generally subside spontaneously and there is some evidence that such therapy may prolong the duration of fecal excretion of the organisms; however, the US Centers for Disease Control and Prevention (CDC),503 American Academy of Pediatrics (AAP),292 Infectious Diseases Society of America (IDSA),570 and others412 recommend anti-infective therapy in individuals with severe Salmonella gastroenteritis and in those who are at increased risk of invasive disease. These individuals at increased risk include infants younger than 3–6 months of age; individuals older than 50 years of age; individuals with hemoglobinopathies, severe atherosclerosis or valvular heart disease, prostheses, uremia, chronic GI disease, or severe colitis; and individuals who are immunocompromised because of malignancy, immunosuppressive therapy, HIV infection, or other immunosuppressive illness.292 412 503 570

When an anti-infective is considered necessary in an individual with Salmonella gastroenteritis, CDC, AAP, IDSA, and others recommend ceftriaxone, cefotaxime, a fluoroquinolone (should be used in children only if the benefits outweigh the risks and no other alternative exists), ampicillin, amoxicillin, co-trimoxazole, or chloramphenicol, depending on the susceptibility of the causative organism.292 412 503 570

Shigella Infections

Ceftriaxone is used for the treatment of shigellosis caused by susceptible Shigella sonnei or S. flexneri292 570 673 674 and is an alternative to fluoroquinolones for treatment of these infections in pediatric patients or when susceptibility is unknown or strains resistant to ampicillin and co-trimoxazole are isolated.292

Vibrio Infections

Cefotaxime is one of several alternatives recommended for the treatment of severe cases of Vibrio parahaemolyticus infection when anti-infective therapy is indicated in addition to supportive care.503 V. parahaemolyticus infections can occur as the result of ingestion of contaminated undercooked or raw fish or shellfish, and some clinicians recommend use of tetracycline, doxycycline, gentamicin, or cefotaxime when treatment is considered necessary.503

Cefotaxime and ceftazidime are considered drugs of choice for the treatment of infections caused by V. vulnificus; these infections can occur as the result of ingesting raw or undercooked seafood (especially raw oysters) or through contamination of a wound with seawater or seafood drippings.503 582 While optimum anti-infective therapy for the treatment of V. vulnificus infections has not been identified,503 582 use of cefotaxime, ceftazidime, tetracycline, or doxycycline is recommended.503 582 710

Yersinia Infections

Cefotaxime is suggested as a possible choice for the treatment of GI infections caused by Yersinia enterocolitica or Y. pseudotuberculosis.503 710 These Yersinia infections usually are self-limited and anti-infective therapy unnecessary; however, AAP, IDSA, and others recommend use of anti-infectives in immunocompromised individuals or for the treatment of severe infections or when septicemia or other invasive disease occurs.292 503 570 GI infections caused by Y. enterocolitica or Y. pseudotuberculosis can occur as the result of ingesting undercooked pork, unpasteurized milk, or contaminated water; infection has occurred in infants whose caregivers handled contaminated chitterlings (raw pork intestines) or tofu.503 Use of co-trimoxazole, an aminoglycoside (amikacin, gentamicin, tobramycin), a fluoroquinolone (e.g., ciprofloxacin), doxycycline, or cefotaxime has been recommended when treatment is considered necessary;503 570 710 combination therapy may be necessary.570 Some clinicians suggest that the role of oral anti-infectives in the management of enterocolitis, pseudoappendicitis syndrome, or mesenteric adenitis caused by Yersinia needs further evaluation.292 675

Intra-abdominal Infections

Cefazolin,708 cefepime,314 708 cefotaxime,332 708 cefoxitin,708 ceftazidime,340 708 ceftriaxone,343 708 and cefuroxime708 are used for the treatment of intra-abdominal infections. The fixed combination of ceftazidime and avibactam728 and the fixed combination of ceftolozane and tazobactam729 are used for the treatment of complicated intra-abdominal tract infections. Certain cephalosporins (e.g., cefotaxime, ceftriaxone, cefotetan, cefoxitin) also are used for the treatment of various obstetric and gynecologic infections, including pelvic inflammatory disease. (See Uses: Pelvic Inflammatory Disease.) Because the prevalence of B. fragilis resistant to cefotetan has been increasing, some experts state that cefotetan is not recommended for empiric treatment of intra-abdominal infections.708

Although monotherapy with cefazolin, ceftriaxone, or cefuroxime is an option for initial empiric treatment of mild to moderate community-acquired biliary tract infections (cholecystitis or cholangitis) and monotherapy with cefoxitin is an option for initial empiric treatment of mild to moderate community-acquired extrabiliary intra-abdominal infections,708 cephalosporins usually should be used in conjunction with metronidazole for initial empiric treatment of community-acquired or healthcare-associated intra-abdominal infections.708

For initial empiric treatment of mild to moderately severe community-acquired extrabiliary intra-abdominal infections in adults, IDSA recommends either monotherapy with cefoxitin, ertapenem, moxifloxacin, tigecycline, or the fixed combination of ticarcillin and clavulanate sodium, or a combination regimen that includes a cephalosporin (cefazolin, cefotaxime, ceftriaxone, cefuroxime) or fluoroquinolone (ciprofloxacin, levofloxacin) in conjunction with metronidazole.708 For initial empiric treatment of high-risk or severe community-acquired extrabiliary intra-abdominal infections in adults (e.g., those with advanced age, immunocompromise, severe physiologic disturbance), IDSA recommends either monotherapy with a carbapenem (doripenem, imipenem, meropenem) or the fixed combination of piperacillin and tazobactam, or a combination regimen that includes either a cephalosporin (cefepime, ceftazidime) or fluoroquinolone (ciprofloxacin, levofloxacin) in conjunction with metronidazole.708 IDSA also recommends ceftazidime or cefepime in conjunction with metronidazole as one of several regimens that can be used for initial empiric treatment of healthcare-associated complicated intra-abdominal infections in adults and recommends a combination regimen that includes a cephalosporin (cefotaxime, ceftriaxone, ceftazidime, cefepime) in conjunction with metronidazole as one of several options that can be used for empiric treatment of community-acquired complicated intra-abdominal infections in pediatric patients.708

For additional information regarding management of intra-abdominal infections, the current IDSA clinical practice guidelines available at [Web] should be consulted.708

Meningitis and Other CNS Infections

IV cefotaxime, ceftazidime, ceftriaxone, and cefuroxime are used in adult or pediatric patients for the treatment of meningitis caused by susceptible H. influenzae, Neisseria meningitidis, or S. pneumoniae; however, cefotaxime or ceftriaxone generally are preferred when a cephalosporin is indicated for the treatment of meningitis caused by these organism. IV cefotaxime and ceftriaxone are used alone or in conjunction with an aminoglycoside for the treatment of meningitis or other CNS infections caused by susceptible Enterobacteriaceae (e.g., E. coli, Klebsiella)392 393 and IV ceftazidime is used in conjunction with an aminoglycoside for the treatment of meningitis caused by susceptible Ps. aeruginosa.393 416

Empiric Treatment of Meningitis

Pending results of CSF culture and in vitro susceptibility testing, the most appropriate anti-infective regimen for empiric treatment of suspected bacterial meningitis should be selected based on results of CSF Gram stain and antigen tests, age of the patient, the most likely pathogen(s) and source of infection, and current patterns of bacterial resistance within the hospital and local community.292 392 393 397 When results of culture and susceptibility tests become available and the pathogen is identified, the empiric anti-infective regimen should be modified (if necessary) to ensure that the most effective regimen is being administered.392 393 There is some evidence that short-term adjunctive therapy with IV dexamethasone may decrease the incidence of audiologic and/or neurologic sequelae in infants and children with H. influenzae meningitis and possibly may provide some benefit in patients with S. pneumoniae meningitis.292 392 393 398 AAP and other clinicians suggest that use of adjunctive dexamethasone therapy should be considered during the initial 2–4 days of anti-infective therapy in infants and children 6–8 weeks of age or older with known or suspected bacterial meningitis and is recommended in those with suspected or proven H. influenzae infection.292 392 393 397 If used, dexamethasone should be initiated before or concurrently with the first dose of anti-infective.292

Bacterial meningitis in neonates usually is caused by S. agalactiae (group B streptococci), Listeria monocytogenes, or aerobic gram-negative bacilli (e.g., E. coli, K. pneumoniae).292 391 392 393 394 397 710 AAP recommends that neonates with suspected bacterial meningitis receive an empiric regimen of IV ampicillin and an aminoglycoside pending results of CSF culture and susceptibility testing.292 An empiric regimen of IV ampicillin and IV cefotaxime or IV ceftazidime with or without gentamicin also is recommended.292 391 392 393 394 397 710 Because frequent use of cephalosporins in neonatal units may result in rapid emergence of resistant strains of some gram-negative bacilli (e.g., Enterobacter cloacae, Klebsiella, Serratia), AAP cautions that cephalosporins should be used for empiric treatment of meningitis in neonates only if gram-negative bacterial meningitis is strongly suspected.292 Consideration should be given to including IV vancomycin in the empiric regimen if S. pneumoniae, enterococci, or Staphylococci is suspected.292 405 406 Because ceftriaxone should be used with caution in neonates who are hyperbilirubinemic (especially those born prematurely), cefotaxime may be the preferred cephalosporin for empiric treatment of meningitis is neonates.292 392 However, because premature, low-birthweight neonates are at increased risk for nosocomial infection caused by staphylococci or gram-negative bacilli, some clinicians suggest that these neonates receive an empiric regimen of IV ceftazidime and IV vancomycin.393

In infants beyond the neonatal stage who are younger than 3 months of age, bacterial meningitis usually is caused by S. agalactiae, L. monocytogenes, H. influenzae, S. pneumoniae, N. meningitidis, or aerobic gram-negative bacilli (e.g., E. coli, K. pneumoniae).292 391 392 393 394 397 The empiric regimen recommended for infants in this age group is IV ampicillin and either IV ceftriaxone or IV cefotaxime.391 392 393 394 397 Consideration should be given to including IV vancomycin in the empiric regimen if S. pneumoniae is suspected.405

In adults and children 3 months through 17 years of age, bacterial meningitis usually is caused by N. meningitidis or S. pneumoniae is N. meningitidis or S. pneumoniae.292 392 393 397 710 An empiric regimen of IV ceftriaxone or IV cefotaxime usually is used for empiric therapy of suspected bacterial meningitis in children 3 months through 17 years of age and in adults 18–50 years of age.292 392 393 394 397 710 Although an empiric regimen of IV ampicillin and IV chloramphenicol can be used as an alternative regimen in children 3 months through 17 years of age,392 most clinicians prefer a cephalosporin regimen unless the drugs are contraindicated.292 392 393 394 397 406 Because of the increasing prevalence of penicillin-resistant S. pneumoniae that also are resistant to or have reduced susceptibility to cephalosporins, AAP and others recommend that the initial empiric cephalosporin regimen include IV vancomycin (with or without rifampin) pending results of in vitro susceptibility tests;292 391 397 403 405 710 vancomycin and rifampin should be discontinued if the causative organism is found to be susceptible to the cephalosporin.292 400 403 405 710 CDC and some clinicians have recommended that vancomycin be added to the empiric regimen in areas where there have been reports of highly penicillin-resistant strains of S. pneumoniae,292 405 407 408 409 410 411 but other clinicians suggest that use of ceftriaxone or cefotaxime in conjunction with vancomycin provides the optimal initial empiric regimen.397 400 While L. monocytogenes meningitis is relatively rare in this age group, the empiric regimen should include ampicillin if L. monocytogenes is suspected.394

In adults older than 50 years of age, bacterial meningitis usually is caused by S. pneumoniae, L. monocytogenes, N. meningitidis, or aerobic gram-negative bacilli, and the empiric regimen recommended for this age group is IV ampicillin given in conjunction with IV cefotaxime or IV ceftriaxone.393 394 Because of the increasing prevalence of penicillin-resistant S. pneumoniae, some clinicians suggest that the empiric regimen also should include IV vancomycin (with or without rifampin).393

Meningitis Caused by Streptococcus pneumoniae

IV ceftriaxone and IV cefotaxime are considered drugs of choice for the treatment of meningitis caused by susceptible S. pneumoniae.292 405 710 Treatment failures have been reported when these cephalosporins were used alone for the treatment of meningitis caused by S. pneumoniae with intermediate or high-level penicillin resistance (i.e., penicillin MIC 0.1 mcg/mL or greater).292 407 409 411 413 434 In addition, strains of S. pneumoniae with reduced susceptibility to cephalosporins have been reported with increasing frequency, and use of cefotaxime or ceftriaxone alone may be ineffective for the treatment of meningitis caused by these strains.396 397 For meningitis caused by S. pneumoniae with high-level resistance to penicillin (i.e., penicillin MIC 8 mcg/mL or greater), IV cefotaxime or IV ceftriaxone is used in conjunction with vancomycin with or without rifampin.710 The prevalence of S. pneumoniae with reduced susceptibility to penicillin and/or cephalosporins varies geographically, and clinicians should be aware of the prevalence and pattern of S. pneumoniae drug resistance in the local community to optimize empiric regimens and initial therapy for serious pneumococcal infections.292 410

Because susceptibility can no longer be assumed, S. pneumoniae isolates should be routinely tested for in vitro susceptibility.292 392 405 407 408 If anti-infective therapy in a patient with meningitis is initiated with an empiric regimen of IV ceftriaxone or IV cefotaxime and IV vancomycin (with or without rifampin) and results of culture and in vitro susceptibility testing indicate that pathogen involved is a strain of S. pneumoniae susceptible to the cephalosporin and susceptible or resistant to penicillin, vancomycin and rifampin can be discontinued and therapy completed using ceftriaxone or cefotaxime alone.292 405 If the isolate is found to have reduced susceptibility to ceftriaxone and cefotaxime and penicillin, the IV cephalosporin and IV vancomycin usually are both continued.292 394 If the patient’s condition does not improve or worsens or results of a second repeat lumbar puncture (performed 24–48 hours after initiation of anti-infective therapy) indicate that the anti-infective regimen has not eradicated or substantially reduced the number of pneumococci in CSF, rifampin probably should be added to the regimen or vancomycin discontinued and replaced with rifampin.292 405 If meningitis is caused by S. pneumoniae highly resistant to ceftriaxone (i.e., MIC 4 mcg/mL or greater), consultation with an infectious disease expert is recommended.292 405

Meningitis Caused by Haemophilus influenzae

IV ceftriaxone and IV cefotaxime are considered drugs of choice for the initial treatment of meningitis caused by susceptible H. influenzae (including penicillinase-producing strains).292 392 393 710 AAP suggests that children with meningitis possibly caused by H. influenzae can receive an initial treatment regimen of ceftriaxone, cefotaxime, or a regimen of ampicillin given in conjunction with chloramphenicol;292 some clinicians prefer ceftriaxone or cefotaxime for the initial treatment of meningitis caused by H. influenzae since these cephalosporins are active against both penicillinase-producing and nonpenicillinase-producing strains. Because of the prevalence of ampicillin-resistant H. influenzae, ampicillin should not be used alone for empiric treatment of meningitis when H. influenzae may be involved.292 The incidence of H. influenzae meningitis in the US has decreased considerably since H. influenzae type b conjugate vaccines became available for immunization of infants.292 394 397

Meningitis Caused by Neisseria meningitidis

While both IV ampicillin and IV penicillin G may be used for the treatment of meningitis caused by N. meningitidis,393 397 AAP and other clinicians suggest that IV penicillin G is the drug of choice for the treatment of these infections and IV ceftriaxone and IV cefotaxime are acceptable alternatives.292 393 Chloramphenicol is recommended for the treatment of N. meningitidis meningitis in patients with a history of anaphylactoid-type hypersensitivity reactions to penicillin.292

Meningitis Caused by Enterobacteriaceae

Some clinicians recommend that meningitis caused by Enterobacteriaceae (e.g., E. coli, K. pneumoniae) be treated with a third generation cephalosporins (i.e., cefotaxime, ceftazidime, ceftriaxone) with or without an aminoglycoside.392 393 Because ceftazidime (but not cefotaxime or ceftriaxone) is effective for the treatment of meningitis caused by Ps. aeruginosa, some clinicians suggest that a regimen of ceftazidime and an aminoglycoside may be preferred for the treatment of meningitis caused by gram-negative bacilli pending results of culture and susceptibility testing.393

Meningitis Caused by Pseudomonas aeruginosa

In patients with meningitis caused by Ps. aeruginosa, most clinicians recommend that therapy be initiated with a regimen of ceftazidime and a parenteral aminoglycoside.393 416 If the patient fails to respond to this regimen, concomitant use of intrathecal or intraventricular aminoglycoside therapy or use of an alternative parenteral anti-infective (e.g., aztreonam, meropenem, a quinolone) should be considered based on results of in vitro susceptibility tests.393 416 When treating pediatric patients with meningitis caused by Ps. aeruginosa or Enterobacteriaceae, consultation with an infectious disease expert may be beneficial.292

Meningitis Caused by Streptococcus agalactiae

For the initial treatment of meningitis or other severe infection caused by S. agalactiae (group B streptococci), a regimen of IV ampicillin or IV penicillin G given in conjunction with an aminoglycoside is recommended.292 393 Some clinicians suggest that IV ampicillin is the drug of choice for the treatment of group B streptococcal meningitis and that an aminoglycoside (IV gentamicin) should be used concomitantly during the first 72 hours until in vitro susceptibility testing is completed and a clinical response if observed; thereafter, ampicillin can be given alone.393

Meningitis Caused by Listeria monocytogenes

The optimal regimen for the treatment of meningitis caused by L. monocytogenes has not been established.292 Cephalosporins are not active against Listeria monocytogenes, an organism that most frequently causes meningitis in neonates or immunocompromised individuals, and the drugs should not be used alone for empiric treatment of meningitis when this organisms may be involved.292 393 394 AAP and other clinicians generally recommend that meningitis or other severe infection caused by L. monocytogenes be treated with a regimen of IV ampicillin with or without an aminoglycoside (usually gentamicin);292 393 414 710 alternatively, a regimen of penicillin G used in conjunction with gentamicin can be used.393 414 In patients hypersensitive to penicillin, the alternative regimen for treatment of meningitis caused by L. monocytogenes is co-trimoxazole.292 710

Brain Abscess and Other CNS Infections

Bacterial brain abscesses and other CNS infections (e.g., subdural empyema, intracranial epidural abscesses) often are polymicrobial and can be caused by gram-positive aerobic cocci, Enterobacteriaceae (e.g., E. coli, Klebsiella), and/or anaerobic bacteria (e.g., Bacteroides, Fusobacterium).292 394 415 The choice of anti-infectives for empiric therapy of these infections should be based on the predisposing condition and site of primary infection.394 415 Some clinicians suggest that the empiric anti-infective regimen in patients who develop the CNS infections after respiratory tract infection (e.g., otitis media, mastoiditis, paranasal sinusitis, pyogenic lung disease) should consist of an appropriate third generation IV cephalosporin (e.g., ceftriaxone, cefotaxime, ceftazidime) given in conjunction with metronidazole; employing one of these cephalosporins rather than a penicillin provides coverage against Haemophilus and facultative anaerobic gram-negative bacteria.394 415 If presence of staphylococci is suspected, a penicillinase-resistant penicillin (e.g., nafcillin, oxacillin) or vancomycin should be added to the empiric regimen.394 415 In patients who develop brain abscess, subdural empyema, or intracranial epidural abscess after trauma or neurosurgery, the empiric regimen should consist of an appropriate third generation IV cephalosporin (e.g., ceftriaxone, cefotaxime, ceftazidime) given in conjunction with a penicillinase-resistant penicillin or vancomycin.394 415 Prolonged anti-infective therapy (e.g., 3–6 weeks or longer) usually is required for the treatment of brain abscess, subdural empyema, or intracranial epidural abscess.394 415

Otitis Media

Acute Otitis Media

Various oral cephalosporins (e.g., cefaclor, cefdinir, cefixime, cefpodoxime proxetil, cefprozil, ceftibuten, cefuroxime axetil, cephalexin) are used for the treatment of acute otitis media (AOM) caused by S. pneumoniae, H. influenzae (including β-lactamase-producing strains), or M. catarrhalis (including β-lactamase-producing strains).307 311 338 339 341 348 349 677 683 Parenteral ceftriaxone also is used for the treatment of AOM caused by S. pneumoniae, H. influenzae (including β-lactamase-producing strains), or M. catarrhalis (including β-lactamase-producing strains).343 683

AOM is one of the most frequently diagnosed bacterial infection in children, and 65–95% of children will have at least one episode of AOM by 3 years of age.436 444 445 449 526 537 543 559 617 622 S. pneumoniae, H. influenzae, and M. catarrhalis are the bacteria most frequently recovered from middle ear fluid of patients with AOM; S. pyogenes and S. aureus also are recovered rarely.348 349 433 448 449 522 526 528 529 551 552 559 568 569 579 617 622 683 In addition, there is evidence that respiratory viruses (e.g., respiratory syncytial virus, rhinoviruses, influenza virus, parainfluenza virus, enteroviruses) may be present either alone or in combination with bacterial pathogens and may play a role in the etiology and pathogenesis of AOM in some patients.433 448 449 533 534 559 586 587 622 683

Diagnosis and Management Strategies for AOM

AAP and American Academy of Family Physicians (AAFP) first issued evidence-based clinical practice guidelines for the diagnosis and management of AOM in 2004.683 In 2013, AAP revised and updated those guidelines after comprehensive reviews of more recent published evidence.683 The 2013 AAP evidence-based clinical practice guidelines provide recommendations for the diagnosis and management of uncomplicated AOM, including recurrent AOM, in children 6 months through 12 years of age and apply only to otherwise healthy children who do not have underlying conditions that may alter the natural course of AOM (e.g., tympanostomy tubes, cleft palate, genetic conditions with craniofacial abnormalities such as Down syndrome, immunodeficiencies, cochlear implants).683 These AAP guidelines should be consulted for additional information on diagnosis and management of AOM.683

Accurate diagnosis of AOM is critical for clinical decision-making since it avoids unnecessary treatment.683 AOM involves the presence of fluid in the middle ear accompanied by a wide spectrum of signs or symptoms of acute local or systemic illness (e.g., otalgia, otorrhea, hearing loss, swelling around the ear, vertigo, nystagmus, tinnitus, fever, irritability, headache, diarrhea, lethargy, anorexia, vomiting) that evolve as the disease progresses.449 528 532 543 559 683 Older children with AOM usually have a history of rapid onset of ear pain,683 but preverbal infants and young children may have mild or nonspecific symptoms that overlap with those of an upper respiratory tract illness.683

Current AAP evidence-based clinical practice guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that clinicians should diagnose AOM in children who present with moderate to severe bulging of the tympanic membrane or new onset of otorrhea not due to acute otitis externa.683 A diagnosis of AOM also should be made in children who present with mild bulging of the tympanic membrane and recent (less than 48 hours) onset of ear pain (holding, tugging, rubbing of the ear in a nonverbal child) or intense erythema of the tympanic membrane.683 These guidelines state that a diagnosis of AOM should not be made in children who do not have middle ear effusion (MEE) based on pneumatic otoscopy and/or tympanometry.683

Current AAP evidence-based clinical practice guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that management of AOM should include an assessment of pain and, if ear pain is present, the clinician should recommend treatment to reduce the pain.683 AOM-associated pain can be substantial during the first few days of illness and often persists longer in young children.683 AAP states that pain management, especially during the first 24 hours of an AOM episode, should be addressed regardless of the use of anti-infectives.683 Treatment for otalgia should be selected based on a consideration of the benefits and risks and, whenever possible, incorporate parent and/or caregiver and patient preference.683 Acetaminophen or ibuprofen are effective for mild to moderate pain, readily available, and usually the mainstay of pain management for AOM.683

Up to 60–80% of cases of AOM resolve spontaneously within 7–14 days,433 436 448 449 523 528 543 594 and routine administration of anti-infectives is not considered necessary for the treatment of all cases of AOM.523 528 532 541 550 588 590 592 593 594 606 683 Some clinicians have recommended that all cases of AOM be treated with an appropriate anti-infective regimen to facilitate resolution of the primary infection and associated symptoms and prevent suppurative complications or other sequelae, and state that judicious use of anti-infectives in the management of otitis media involves accurately diagnosing AOM and distinguishing AOM (which should be treated with anti-infectives) from otitis media with effusion (which usually is not treated with anti-infectives).532 537 539 540 541 543 546 548 549 560 562 591 622 However, for the majority of patients with uncomplicated AOM, anti-infective therapy appears to provide only minimal benefits in terms of resolution of the acute symptoms of infection (e.g., pain) and the proposed benefits of such therapy in terms of time to bacteriologic or clinical resolution of AOM or in terms of long-term consequences of otitis media (e.g., persistence of middle ear effusion, recurrence of AOM, hearing loss, need for adenoidectomy or insertion of tympanostomy tubes, mastoiditis) have never been substantiated in well-designed, placebo-controlled studies.550 588 592 593 594 595 683 In addition, there is evidence that overuse of anti-infectives, including overuse in the treatment of AOM, contributes to emergence of resistant bacteria (e.g., multidrug-resistant S. pneumoniae).588 592 594 596 615 Based on these considerations, many clinicians now recommend a management strategy for AOM that involves use of symptomatic care with analgesics and close observation via telephone contact or office visits for the majority of patients with uncomplicated AOM and use of anti-infectives only in those who do not have symptomatic improvement within 24–72 hours after diagnosis and in those who appear least likely to have spontaneous resolution and most likely to have poor outcomes (e.g., more acutely ill, those with 3 or more episodes of AOM in the past 18 months, history of serous otitis or tympanostomy tubes).588 595 606 683

Current AAP evidence-based clinical practice guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age include an initial management option of observation with close follow-up without initial use of anti-infectives in certain selected children with uncomplicated AOM based on age, illness severity, and assurance of follow-up.683 The recommendation for initial observation with close follow-up in select children provides an opportunity for the patient to improve without anti-infectives and is based on results of randomized, controlled studies with limitations and consideration of the benefits and risks of such a strategy.683

Current AAP guidelines state that anti-infective treatment should be initiated in children 6 months of age or older who have AOM (bilateral or unilateral) with severe signs or symptoms (i.e., moderate or severe otalgia, otalgia for at least 48 hours, or temperature 39°C or higher) and in children 6 through 23 months of age who have nonsevere bilateral AOM without signs or symptoms (i.e., mild otalgia for less than 48 hours and temperature less than 39°C).683 However, these guidelines state that a management strategy of either initiation of anti-infective treatment or observation with close follow-up can be used in children 6 through 23 months of age who have nonsevere unilateral AOM without severe signs or symptoms (i.e., mild otalgia for less than 48 hours, temperature less than 39°C) and in children 24 months of age or older with nonsevere AOM (bilateral or unilateral) without severe signs or symptoms (i.e., mild otalgia for less than 48 hours, temperature less than 39°C).683 The strategy of observation with close follow-up should be based on joint decision-making between the clinician and the parent and/or caregiver and must include a mechanism that ensures follow-up and initiation of anti-infective therapy if AOM worsens or fails to improve within 48–72 hours after symptom onset.683

If the initial management strategy was observation with close follow-up, anti-infective therapy should be initiated if symptoms worsen or there is no improvement within 48–72 hours after onset of symptoms.683 If the initial management strategy was anti-infective treatment, consideration should be given to changing the anti-infective regimen if symptoms worsen or fail to respond within 48–72 hours after initiation of treatment.683 (See Anti-infectives for AOM after Initial Treatment Failure under Uses: Otitis Media.)

After the patient has shown clinical improvement, follow-up is based on the usual clinical course of AOM.683 Persistent MEE is common after resolution of acute symptoms of AOM and should not be viewed as requiring anti-infective treatment.683 (See Otitis Media with Effusion under Uses: Otitis Media.)

Anti-infectives for Initial Treatment of AOM

When anti-infectives are indicated for treatment of AOM, the initial anti-infective agent usually is selected empirically based on efficacy against the most probable bacterial pathogens.526 528 537 543 557 560 568 619 Other considerations in the choice of an anti-infective for initial empiric treatment of AOM include pharmacokinetic data related to distribution of the drug into middle ear fluid, compliance issues related to patient acceptance of dosage formulation and dosage schedule, adverse effects profiles, and cost considerations;448 449 522 523 528 529 531 565 571 574 577 619 drug susceptibility patterns in the local community can be considered, but local surveillance data are not necessarily representative of AOM isolates found in otherwise healthy patients.522

Amoxicillin usually is considered the drug of first choice for initial empiric treatment of AOM, unless the infection is suspected of being caused by β-lactamase-producing bacteria resistant to the drug, in which case amoxicillin and clavulanate potassium is recommended.292 330 331 334 336 391 433 436 448 449 522 524 560 565 574 619 622 683 The fact that multidrug-resistant S. pneumoniae are being reported with increasing frequency should be considered when selecting an anti-infective agent for empiric treatment of AOM.522 527 538 553 563 564 565 569 583 584 619 622 633 However, AAP, AAFP, CDC, and others state that, despite the increasing prevalence of multidrug-resistant S. pneumoniae and presence of β-lactamase-producing H. influenzae or M. catarrhalis in many communities, amoxicillin remains the anti-infective of first choice for treatment of uncomplicated AOM since amoxicillin is highly effective, has a narrow spectrum of activity, is well distributed into middle ear fluid, is well tolerated, has an acceptable taste, and is inexpensive.292 522 527 528 537 538 557 560 561 615 619 622 683 Amoxicillin (when given in dosages of 80–90 mg/kg daily) usually is effective in the treatment of AOM caused by S. pneumoniae, including infections involving strains with intermediate resistance to penicillins, and also usually is effective in the treatment of AOM caused by most strains of H. influenzae.522 527 560 683 Because S. pneumoniae is the most frequent cause of AOM (25–50% of cases) and because AOM caused by S. pneumoniae is more likely to be severe and less likely to resolve spontaneously than AOM caused by H. influenzae or M. catarrhalis, it has been suggested that it may be more important to choose an empiric anti-infective based on its activity against S. pneumoniae rather than its activity against other possible pathogens.449 522 527 538

Various other anti-infectives, including oral cephalosporins (cefaclor, cefdinir, cefixime, cefpodoxime proxetil, cefprozil, ceftibuten, cefuroxime axetil, cephalexin), parenteral ceftriaxone, oral macrolides (azithromycin, clarithromycin, fixed combination of erythromycin and sulfisoxazole), oral co-trimoxazole, and oral loracarbef, have been used in the treatment of AOM.292 330 333 334 336 391 433 436 448 449 527 557 565 574 610 616 619 622 448 522 683 These anti-infectives vary in their efficacy against AOM pathogens.683

Current AAP evidence-based guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that high-dose amoxicillin (80–90 mg/kg daily in 2 divided doses) should be used for initial treatment when a decision has been made to use anti-infective therapy and the child has not received amoxicillin within the past 30 days or does not have concurrent purulent conjunctivitis or is not allergic to penicillin.683 These guidelines state that high-dose amoxicillin and clavulanate (90 mg/kg of amoxicillin and 6.4 mg/kg of clavulanate daily in 2 divided doses) should be used if the child received amoxicillin within the past 30 days or has concurrent purulent conjunctivitis or has a history of recurrent AOM unresponsive to amoxicillin.683 These AAP guidelines state that the preferred alternatives for initial treatment of AOM in penicillin-allergic patients are oral cephalosporins (cefdinir, cefpodoxime proxetil, cefuroxime axetil) or parenteral ceftriaxone.683

Results of controlled clinical studies indicate that 10-day regimens of most oral anti-infectives used in the empiric treatment of AOM are equally effective, and there is no evidence that the overall response rate to anti-infectives with a broader spectrum of activity (e.g., second and third generation cephalosporins) is any better than that reported with amoxicillin or amoxicillin and clavulanate potassium.443 449 527 548 555 556 557 566 567 573 609 610 619 Of the currently available oral cephalosporins, cefaclor,307 cefdinir,311 cefixime,677 cefpodoxime proxetil,338 cefprozil,339 ceftibuten,341 and cefuroxime axetil348 576 have been used effectively for the treatment of AOM in pediatric patients. However, there is evidence that some cephalosporins (e.g., cefaclor, cefprozil) may be less effective than some other available agents for the treatment of AOM when β-lactamase-producing bacteria are present330 331 333 334 336 337 339 433 585 619 621 622 and some (e.g., cefixime, ceftibuten) may be less effective than some other available agents for the treatment of AOM when S. pneumoniae with reduced susceptibility to penicillin are present.341 448 538 584 585 615 622 677

Duration of Initial Treatment of AOM

The optimal duration of therapy for AOM is uncertain.683 Anti-infectives traditionally have been administered for 7–10 days for the treatment of AOM,292 543 546 557 578 but shorter durations of treatment also have been used.540 542 543 546 The 10-day regimen was derived from the duration of treatment for S. pyogenes pharyngitis and tonsillitis.683 Although there is some evidence from controlled clinical studies in pediatric patients with AOM that the clinical response rate to 5-day regimens of certain oral cephalosporins (e.g., cefaclor,446 cefdinir,311 cefpodoxime proxetil,338 441 442 443 cefprozil,445 447 cefuroxime axetil581 633 ) is similar to that of 10-day regimens of oral cephalosporins, amoxicillin, or amoxicillin and clavulanate potassium,444 446 581 633 such regimens appear to be less effective than 10-day regimens in children younger than 2 years of age.683

Current AAP evidence-based guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that a 10-day regimen of an appropriate oral anti-infective is recommended for the treatment of AOM in children younger than 2 years of age and in those with severe symptoms.683 These guidelines state that a 7-day regimen of an appropriate oral anti-infective may be as effective as a 10-day regimen in children 2–5 years of age with mild to moderate AOM, and a 5- to 7-day regimen of an appropriate oral anti-infective may be adequate in children 6 years of age or older with mild to moderate AOM.683

Other clinicians suggest that while 5-day regimens can be considered for adults and children 2 years of age and older with mild, uncomplicated AOM,292 543 further study is needed to more fully evaluate efficacy of short-term regimens in infants and young children since studies to date have included only a limited number of children younger than 2 years of age.406 436 444 446 447 540 541 542 543 683 These clinicians state that short-term anti-infective regimens (i.e., 5 days or less) may not be appropriate for the treatment of AOM in children younger than 2 years of age or for patients with underlying disease, craniofacial abnormalities, recurrent or persistent AOM, or perforated tympanic membranes and spontaneous purulent drainage.292 406 436 444 447 540 541 542 543 546

Parenteral Ceftriaxone for Treatment of AOM

Parenteral ceftriaxone has been shown to be effective for initial or repeat treatment of AOM,343 421 535 613 614 683 and is a good choice when the patient has persistent vomiting or cannot otherwise tolerate an oral regimen.683 The manufacturer recommends a single dose of IM ceftriaxone for initial treatment of AOM, but cautions that the potential advantages of this single-dose parenteral regimen should be balanced against its potentially lower clinical cure rate compared with a 10-day oral anti-infective regimen.343 AAP states that either a 1- or 3-day regimen of parenteral ceftriaxone can be used for initial treatment of AOM, but cautions that there is some evidence that more than a single dose of the drug may be required to prevent recurrence of middle ear infections within 5–7 days after the initial dose.683 A 3-day regimen of parenteral ceftriaxone is recommended for retreatment in patients who fail to respond to initial treatment with other anti-infectives.525 613 683

The single-dose IM ceftriaxone regimen offers some practical advantages over 5- to 10-day oral anti-infective regimens since it provides a more convenient dosing schedule, ensures compliance, and can be administered to patients who have nausea and vomiting;421 422 423 604 614 however, ceftriaxone may be more costly than oral regimens and the drug has a spectrum of activity that is broader than necessary for the treatment of AOM.602 603 Some clinicians suggest that further study of the single-dose ceftriaxone regimen is needed to more fully assess the bacteriologic eradication rate, long-term efficacy, and rate of relapse, and to determine whether the single-dose regimen contributes to emergence of resistant organisms.422 423 424 602

Anti-infectives for AOM after Initial Treatment Failure

Consideration can be given to changing the anti-infective regimen in children who do not have clinical improvement within 48–72 hours after the initial anti-infective regimen is started.683 Current AAP evidence-based clinical practice guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that patients who fail to respond to an initial regimen of high-dose amoxicillin (80–90 mg/kg daily in 2 divided doses) should be retreated with high-dose amoxicillin and clavulanate (90 mg/kg of amoxicillin and 6.4 mg/kg of clavulanate daily in 2 divided doses).683 Those who fail to respond to an initial regimen of high-dose amoxicillin and clavulanate potassium or an initial regimen of an appropriate oral cephalosporin (cefdinir, cefpodoxime proxetil, cefuroxime axetil) should be treated with parenteral ceftriaxone (50 mg/kg daily for 3 days).683

There is evidence that a 3-day regimen of IM ceftriaxone (50 mg/kg once daily) can be effective for the treatment of persistent or recurrent AOM in pediatric patients 3 months of age or older with infections that failed to respond to treatment with other anti-infectives (e.g., amoxicillin, amoxicillin and clavulanate potassium, cefaclor, cefuroxime axetil).535 613 A 3-day regimen of ceftriaxone has been shown to be more effective than a 1-day regimen for retreatment in patients who fail to respond to initial treatment with another anti-infective.683 The 3-day ceftriaxone regimen has been effective for the treatment of persistent or relapsing otitis media caused by H. influenzae, M. catarrhalis, S. pyogenes, or penicillin-susceptible S. pneumoniae; however, treatment failures have been reported when the causative agent was S. pneumoniae with reduced susceptibility to penicillin.535 613

Clindamycin (30–40 mg/kg daily in 3 divided doses) can be used (with or without a third generation cephalosporin) as an alternative for the treatment of AOM in patients who fail to respond to an initial anti-infective regimen.683 Although clindamycin may be effective for penicillin-resistant S. pneumoniae, it may not be effective against multidrug-resistant S. pneumoniae and lacks efficacy against H influenzae.683 If clindamycin is used for retreatment, concomitant use of an anti-infective active against H. influenzae and M. catarrhalis (e.g., cefdinir, cefixime, cefuroxime) should be considered.683

Because of reported resistance in S. pneumoniae, AAP states that co-trimoxazole and the fixed combination of erythromycin and sulfisoxazole should not be used as alternatives for the treatment of AOM in patients who fail to improve while receiving amoxicillin.683

Primary treatment failure of AOM occurs most frequently in children younger than 2 years of age.525 536 544 While primary treatment failure and persistent AOM may be the result of infection with bacteria resistant to the anti-infective administered (e.g., penicillin-resistant S. pneumoniae, β-lactamase-producing H. influenzae),525 529 536 537 544 552 559 565 many cases appear to be related to other factors since results of tympanocentesis indicate that the causative organism(s) often are susceptible in vitro to the primary treatment regimen or, in some cases, no bacteria are isolated.536 544 545 559 Patients with AOM who fail to respond to an initial anti-infective regimen often also fail to respond to a subsequent regimen, regardless of the anti-infective used.522 545

If AOM persists after a series of anti-infective regimens, tympanocentesis should be considered and culture of middle ear fluid performed to make a bacteriologic diagnosis and obtain in vitro susceptibility test results.683 If tympanocentesis is not available, a regimen of oral clindamycin with or without an anti-infective to provide coverage against H. influenzae and M. catarrhalis (e.g., cefdinir, cefixime, cefuroxime) may be considered.683 Consultation with a pediatric medical subspecialist (e.g., otolaryngologist) for possible tympanocentesis, drainage, and culture and consultation with an infectious disease expert before use of unconventional anti-infectives should be considered.683

Recurrent AOM

Current AAP evidence-based clinical practice guidelines for the diagnosis and management of uncomplicated AOM in children 6 months through 12 years of age state that anti-infective prophylaxis should not be prescribed to reduce the frequency of episodes of AOM in children with recurrent AOM.683 Recurrent AOM is defined as 3 or more episodes of AOM within a 6-month period or 4 or more episodes of AOM within a 12-month period that includes at least 1 episode in the preceding 6 months.683 About 50% of children younger than 2 years of age who are treated for AOM will have a recurrence within 6 months.683 Winter season, male gender, and passive exposure to tobacco smoke have been associated with an increased likelihood of AOM recurrence.683 Other risk factors for recurrence include AOM symptoms lasting more than 10 days, a family history of the infection, group day-care outside the home during the first 2 years of life, and use of bottles or pacifiers.449 543 617 683 There is some evidence that breastfeeding for at least 4–6 months reduces episodes of AOM and recurrent AOM.449 617 683

Anti-infectives (e.g., amoxicillin, sulfisoxazole) have been administered as long-term prophylaxis or suppressive therapy in children with recurrent AOM in an attempt to prevent recurrence292 543 547 612 617 622 645 or have been administered intermittently as prophylaxis at the first sign of an upper respiratory tract infection in children with a history of recurrent AOM.541 543 612 617 Although there is some evidence that anti-infective prophylaxis may decrease the incidence of new symptomatic episodes of AOM in some children with a history of recurrent AOM,449 527 532 543 546 547 559 683 such prophylaxis is not routinely recommended.292 532 543 546 617 645 683 Results of a meta-analysis indicate that use of anti-infective prophylaxis results in an average decrease of only 0.11 episodes of AOM per patient per month (slightly more than 1 episode per year).547 In addition, anti-infective prophylaxis does not provide any long-lasting benefit since any decrease in AOM episodes occurs only while prophylaxis is being given.683 AAP states that anti-infective prophylaxis is not appropriate for children with long-term MEE or for children with infrequent episodes of AOM.683 The small reduction in frequency of AOM must be weighed against the cost and potential adverse effects of anti-infective prophylaxis (e.g., allergic reactions, GI effects such as diarrhea)683 and concerns that prophylaxis may promote emergence of resistant bacteria, including multidrug-resistant S. pneumoniae, or alter nasopharyngeal flora and foster colonization with resistant bacteria.292 527 532 541 546 612 617 683

Otitis Media with Effusion

Anti-infectives are not usually recommended for management of otitis media with effusion (OME).292 448 541 543 546 547 684 OME involves liquid in the middle ear (MEE) without signs and symptoms of acute ear infection.683 684 OME may occur as an inflammatory response after an episode of AOM or may occur spontaneously as a consequence of eustachian tube dysfunction.683 684 About 60–70% of children have MEE present 2 weeks after successful treatment of AOM and 10–25% still have MEE at 3 months.683 It is important to differentiate OME from AOM.683 684 Management of OME usually involves observation and close monitoring until resolution.683 684 Anti-infective treatment is not usually indicated because OME does not represent an acute infectious process.683 684 The AAFP, American Academy of Otolaryngology-Head and Neck Surgery, and AAP Subcommittee on Otitis Media with Effusion have issued evidence-based clinical practice guidelines regarding diagnosis and management of OME in children 2 months to 12 years of age (with or without developmental disabilities or underlying conditions that predispose to OME and its sequelae).684 These guidelines should be consulted for information on diagnosis and management of OME.684

Chronic Suppurative Otitis Media without Cholesteatoma

Ceftazidime has been used with some success in the treatment of chronic suppurative otitis media (CSOM) without cholesteatoma.598 600 601 CSOM is defined as chronic infection of the middle ear and mastoid associated with tympanic membrane perforation and otorrhea lasting more than 6 weeks, and may occur as the result of unresolved AOM and/or eustachian tube dysfunction.598 608 622 The most common bacteria reported in patients with CSOM are Ps. aeruginosa, Klebsiella, S. aureus, S. epidermidis, and anaerobic bacteria, including Bacteroides, Prevotella, Peptostreptococcus, and Peptococcus.554 598 599 600 608 622 While ceftazidime has been effective in the treatment of CSOM when gram-negative bacteria were involved, it has been ineffective when gram-positive bacteria (e.g., S. aureus) were involved.598 600 Because CSOM often is a mixed aerobic-anaerobic infection, anti-infectives usually used in the treatment of AOM or OME would be ineffective.554 622 Topical anti-infectives (e.g., ciprofloxacin otic suspension, ofloxacin otic solution, gentamicin) used in conjunction with daily aural toilet can be effective for the treatment of uncomplicated CSOM; more severe or persistent infections require treatment with a parenteral anti-infective (e.g., ceftazidime, clindamycin, ciprofloxacin, gentamicin, ticarcillin, ticarcillin disodium and clavulanate potassium).554 618 622 630

Otitis Externa

Ceftazidime has been effective when used in the treatment of malignant otitis externa caused by Ps. aeruginosa.605 632 Bacterial otitis externa usually is caused by Ps. aeruginosa or S. aureus.631 632 Although acute bacterial otitis externa localized in the external auditory canal may be effectively treated using topical anti-infectives (e.g., ciprofloxacin otic suspension, ofloxacin otic solution), malignant otitis externa is an invasive, potentially life-threatening infection, especially in immunocompromised patients such as those with diabetes mellitus or human immunodeficiency virus (HIV) infection, and requires prompt diagnosis and long-term treatment with parenteral anti-infectives (e.g., ceftazidime and/or ciprofloxacin).605 632

Pharyngitis and Tonsillitis

Oral cephalosporins (e.g., cefaclor, cefadroxil, cefdinir, cefditoren pivoxil, cefixime, cefpodoxime proxetil, cefprozil, ceftibuten, cefuroxime axetil, cephalexin) are used for the treatment of pharyngitis and tonsillitis caused by S. pyogenes (group A β-hemolytic streptococci).292 307 308 311 338 339 341 348 375 663 580 677 Although cephalosporins usually are effective in eradicating S. pyogenes from the nasopharynx, efficacy of the drugs in the subsequent prevention of rheumatic fever remains to be established.307 308 311 338 339 341 348 663 677

Selection of an anti-infective for the treatment of S. pyogenes pharyngitis and tonsillitis should be based on the drug’s spectrum of activity, bacteriologic and clinical efficacy, potential adverse effects, ease of administration, patient compliance, and cost.292 375 433 435 436 580 No regimen has been found to date that effectively eradicates group A β-hemolytic streptococci in 100% of patients.375

Because the drugs have a narrow spectrum of activity, are inexpensive, and generally are effective with a low frequency of adverse effects, AAP,292 IDSA,580 American Heart Association (AHA),375 and other experts678 recommend a penicillin regimen (i.e., 10 days of oral penicillin V or oral amoxicillin or a single dose of IM penicillin G benzathine) as the treatment of choice for S. pyogenes pharyngitis and tonsillitis and prevention of initial attacks (primary prevention) of rheumatic fever. Other anti-infectives (e.g., oral cephalosporins, oral macrolides, oral clindamycin) are recommended as alternatives in penicillin-allergic patients.292 375 378 433 435 436 580 678

A 10-day regimen of an appropriate oral cephalosporin is recommended for the treatment of S. pyogenes pharyngitis and tonsillitis in most penicillin-allergic patients; however, cephalosporins should be avoided in those with a history of immediate (anaphylactic-type) penicillin hypersensitivity.375 580 If an oral cephalosporin is used, a 10-day regimen of a first generation cephalosporin (cefadroxil, cephalexin) is preferred instead of other cephalosporins with broader spectrums of activity (e.g., cefaclor, cefdinir, cefixime, cefpodoxime proxetil, cefuroxime).375 580 The broader spectrum cephalosporins offer no advantages over penicillins or first generation cephalosporins since they generally are more expensive and more likely to result in resistant flora.375 580

Although there is some evidence that bacteriologic and clinical cure rates reported with 10-day regimens of certain oral cephalosporins (e.g., cefaclor, cefadroxil, cefdinir, cefixime, cefpodoxime proxetil, cefprozil, cefuroxime axetil, ceftibuten, cephalexin) may be slightly higher than those reported with the 10-day oral penicillin V regimen,375 377 378 379 381 382 433 435 436 438 analysis of data from the 10-day cephalosporin studies suggests that the difference in eradication rates may have been due to a higher rate of eradication of carriers inadvertently included in the study population.375 In addition, although there is some evidence that a shorter duration of therapy with certain oral cephalosporins (e.g., a 5-day regimen of cefadroxil, cefdinir, cefixime, or cefpodoxime proxetil or a 4- or 5-day regimen of cefuroxime axetil) achieves bacteriologic and clinical cure rates equal to or greater than those achieved with the traditional 10-day oral penicillin V regimen,375 378 382 436 438 439 440 580 AHA and IDSA state that use of cephalosporin regimens administered for 5 days or less for the treatment of S. pyogenes pharyngitis and tonsillitis cannot be recommended at this time.375 580

Respiratory Tract Infections

Community-acquired Pneumonia

Some oral cephalosporins (cefdinir, cefpodoxime proxetil, cefprozil, cefuroxime axetil) and some parenteral cephalosporins (cefepime, cefotaxime, ceftaroline fosamil, ceftriaxone) are used in the treatment of community-acquired pneumonia (CAP) caused by susceptible bacteria.311 680 703

Initial treatment of CAP generally involves use of an empiric anti-infective regimen selected based on the most likely pathogens and local susceptibility patterns; therapy may then be changed (if possible) to provide a more specific regimen (pathogen-directed therapy) based on results of in vitro culture and susceptibility testing.680 The most appropriate empiric regimen varies depending on the severity of illness at the time of presentation and whether outpatient treatment or hospitalization in or out of an intensive care unit (ICU) is indicated and the presence or absence of cardiopulmonary disease and other modifying factors that increase the risk of certain pathogens (e.g., penicillin- or multidrug-resistant S. pneumoniae, enteric gram-negative bacilli, Ps. aeruginosa).680

For both outpatients and inpatients, most experts recommend that an empiric regimen for treatment of CAP include an anti-infective active against S. pneumoniae since this organism is the most commonly identified cause of bacterial pneumonia and causes more severe disease than many other common CAP pathogens.680 682 Although monotherapy may be appropriate for empiric treatment of CAP in many outpatients and some inpatients, a combination regimen is always indicated for empiric treatment of CAP in patients requiring treatment in an ICU.680 Such patients require an empiric regimen that covers S. pneumoniae, Legionella, H. influenzae, Mycoplasma pneumoniae, Chlamydophila pneumoniae (formerly Chlamydia pneumoniae), and relevant gram-negative bacilli.680

Outpatient Regimens for CAP

Pathogens most frequently involved in CAP infections in outpatients include S. pneumoniae, M. pneumoniae, C. pneumoniae, respiratory viruses, and H. influenzae (especially in cigarette smokers).680

For empiric outpatient treatment of CAP in previously healthy individuals without risk factors for drug-resistant S. pneumoniae, IDSA and the American Thoracic Society (ATS) recommend monotherapy with a macrolide (azithromycin, clarithromycin, erythromycin) or, alternatively, doxycycline.680

For empiric outpatient treatment of CAP in patients with risk factors for drug-resistant S. pneumoniae (e.g., comorbidities such as chronic heart, lung, liver, or renal disease, diabetes mellitus, alcoholism, malignancies, asplenia, immunosuppression, history of anti-infective treatment within the last 3 months), ATS and IDSA recommend monotherapy with a fluoroquinolone with enhanced activity against S. pneumoniae (gemifloxacin, levofloxacin, moxifloxacin) or, alternatively, a combination regimen that includes a β-lactam active against S. pneumoniae (high-dose amoxicillin or fixed combination of amoxicillin and clavulanic acid or, alternatively, ceftriaxone, cefpodoxime proxetil, or cefuroxime) given in conjunction with a macrolide (azithromycin, clarithromycin, erythromycin) or, alternatively, given in conjunction with doxycycline.680

Inpatient Regimens for CAP

Pathogens most frequently involved in CAP infections in non-ICU inpatients include S. pneumoniae, M. pneumoniae, C. pneumoniae, H. influenzae, Legionella, and respiratory viruses.680 Patients with severe CAP admitted into the ICU usually have infections caused by S. pneumoniae, S. aureus, Legionella, gram-negative bacilli, or H. influenzae.680 Factors that increase the risk of Enterobacteriaceae or Ps. aeruginosa infection in CAP patients include severe CAP requiring treatment in an ICU, structural lung disease (bronchiectasis), severe chronic obstructive pulmonary disease (COPD), smoking, alcoholism, chronic corticosteroid therapy, and frequent anti-infective therapy.680 Coverage against anaerobic bacteria usually is indicated only in classic aspiration pleuropulmonary syndrome in patients who had loss of consciousness as a result of alcohol or drug overdosage or after seizures in patients with concomitant gingival disease or esophageal motility disorders.680

For empiric inpatient treatment of CAP in non-ICU patients, IDSA and ATS recommend monotherapy with a fluoroquinolone with enhanced activity against S. pneumoniae (gemifloxacin, levofloxacin, moxifloxacin) or, alternatively, a combination regimen that includes a β-lactam (usually cefotaxime, ceftriaxone, or ampicillin) given in conjunction with a macrolide (azithromycin, clarithromycin, erythromycin) or doxycycline.680

For empiric treatment of CAP in ICU patients when Pseudomonas and methicillin-resistant S. aureus (MRSA; also known as oxacillin-resistant S. aureus or ORSA) are not suspected, IDSA and ATS recommend a combination regimen that includes a β-lactam (cefotaxime, ceftriaxone, fixed combination of ampicillin and sulbactam) given in conjunction with either azithromycin or a fluoroquinolone (moxifloxacin, gemifloxacin, levofloxacin).680 If Pseudomonas is suspected, IDSA and ATS recommend an empiric combination regimen that includes an antipneumococcal, antipseudomonal β-lactam (cefepime, imipenem, meropenem, fixed combination of piperacillin and tazobactam) given in conjunction with a fluoroquinolone (ciprofloxacin, levofloxacin); a combination regimen that includes one of these antipseudomonal β-lactams, an aminoglycoside, and azithromycin; or a combination regimen that includes one of these antipseudomonal β-lactams, an aminoglycoside, and an antipneumococcal fluoroquinolone.680 If Ps. aeruginosa has been identified by appropriate microbiologic testing, the preferred treatment regimen is an antipseudomonal β-lactam (cefepime, ceftazidime, aztreonam, imipenem, meropenem, piperacillin, ticarcillin) given in conjunction with ciprofloxacin, levofloxacin, or an aminoglycoside and the preferred alternative regimen is an aminoglycoside given in conjunction with ciprofloxacin or levofloxacin.680 If community-acquired MRSA may be involved, vancomycin or linezolid should be included in the initial empiric regimen.680

Inpatient treatment of CAP is initiated with a parenteral regimen, although therapy may be changed to an oral regimen if the patient is improving clinically, is hemodynamically stable, able to ingest drugs, and has a normally functioning GI tract.680 CAP patients usually have a clinical response within 3–7 days after initiation of therapy and a switch to oral therapy generally can be made during this period.680

Acute Sinusitis

Oral cefdinir,311 cefixime,192 193 cefpodoxime proxetil,338 cefprozil,339 and cefuroxime axetil348 are used for the treatment of acute maxillary sinusitis caused by susceptible bacteria (e.g., S. pneumoniae, H. influenzae, M. catarrhalis). Parenteral ceftriaxone and cefotaxime are recommended as alternatives for initial treatment of severe bacterial sinusitis.192 193

When anti-infective therapy is indicated for the treatment of acute bacterial sinusitis, IDSA recommends amoxicillin and clavulanate potassium and AAP recommends either amoxicillin or amoxicillin and clavulanate potassium as the drug of choice for initial empiric treatment.192 193 Because of variable activity against S. pneumoniae and H. influenzae, IDSA no longer recommends second or third generation oral cephalosporins for empiric monotherapy of sinusitis in adults or children.192 If an oral cephalosporin is used as an alternative for empiric treatment of acute bacterial sinusitis in children (e.g., in penicillin-allergic individuals), IDSA and AAP recommend a combination regimen that includes a third generation cephalosporin (cefixime or cefpodoxime) and clindamycin (or linezolid).192 193 In children who are vomiting, unable to tolerate oral therapy, or unlikely to adhere to the initial doses, treatment of acute sinusitis can be initiated with a single dose of parenteral ceftriaxone and then switched to an oral regimen if clinical improvement is observed at 24 hours.192 193 For patients with severe bacterial sinusitis requiring hospitalization, parenteral ceftriaxone and cefotaxime are preferred alternatives for initial treatment in penicillin-allergic patients who cannot receive amoxicillin and clavulanate potassium.192 193

Septicemia

Cefotaxime,332 ceftazidime,340 ceftriaxone,343 cefazolin,310 and cefepime710 are used parenterally for the treatment of bacteremia/septicemia caused by susceptible bacteria (e.g., S. aureus, S. pneumoniae, H. influenzae, E. coli, K. pneumoniae, Serratia).

The choice of anti-infective for the treatment of sepsis syndrome should be based on the probable source of infection, causative organism, immune status of the patient, and local patterns of bacterial resistance.710 Some experts state that, although certain parenteral cephalosporins (cefepime, cefotaxime, ceftriaxone, ceftazidime) can be used for the treatment of gram-negative sepsis, ceftazidime is less active than the other cephalosporins against gram-positive cocci and most cephalosporins (except cefepime and ceftazidime) have limited activity against Ps. aeruginosa.710

For initial treatment of life-threatening sepsis in adults, some clinicians recommend that a third or fourth generation cephalosporin (cefepime, cefotaxime, ceftazidime, ceftriaxone), the fixed combination of piperacillin and tazobactam, or a carbapenem (doripenem, imipenem, meropenem) be used in conjunction with vancomycin; some experts also suggest including an aminoglycoside or fluoroquinolone during the initial few days of treatment.710

Urinary Tract Infections

Some oral cephalosporins (cefaclor, cefadroxil, cefixime, cefpodoxime proxetil, cefuroxime axetil, cephalexin) and some parenteral cephalosporins (cefazolin, cefepime, ceftazidime, ceftriaxone, cefuroxime) are used for the treatment of uncomplicated and/or complicated urinary tract infections caused by susceptible bacteria.307 308 310 314 338 340 343 347 348 349 677 The fixed combination of ceftazidime and avibactam728 and the fixed combination of ceftolozane and tazobactam729 are used for the treatment of complicated urinary tract infections.

Chancroid

Ceftriaxone is used for the treatment of genital ulcers caused by H. ducreyi (chancroid), and is considered a drug of choice for the treatment of this infection.344

Gonorrhea and Associated Infections

Cefoxitin,344 cefotaxime,344 ceftriaxone,292 343 344 and cefuroxime347 are used parenterally and cefixime,292 344 677 cefpodoxime proxetil,292 338 and cefuroxime axetil348 have been used orally for the treatment of uncomplicated gonorrhea caused by susceptible Neisseria gonorrhoeae. Cefoxitin, cefotaxime,292 ceftriaxone,344 and cefuroxime347 also have been used parenterally for the treatment of disseminated gonorrhea and other gonococcal infections.

Ceftriaxone is considered the drug of choice for the treatment of most N. gonorrhoeae infections, including uncomplicated gonococcal infections of the cervix, urethra, rectum, and pharynx; disseminated gonococcal infections; gonococcal conjunctivitis; gonococcal meningitis and endocarditis; gonococcal epididymitis or proctitis in adults and adolescents; gonococcal ophthalmia neonatorum; and uncomplicated gonorrhea or disseminated gonococcal infections in neonates and children.292 344 Ceftriaxone also is used in conjunction with other agents for empiric anti-infective prophylaxis in sexual assault victims.344 CDC states that ceftriaxone is the most effective cephalosporin for the treatment of gonorrhea716 and, unlike most other drugs, is effective for the treatment of gonorrhea at all sites, including cervical, urethral, rectal, and pharyngeal gonococcal infections.344 Other parenteral or oral cephalosporins do not offer any advantages over ceftriaxone.344

N. gonorrhoeae with reduced susceptibility to ceftriaxone and/or cefixime or other cephalosporins have been reported with increasing frequency during the last decade in the US and elsewhere (e.g., Asia, Europe, Canada);344 716 717 718 719 720 721 722 723 724 725 726 727 some treatment failures have been reported.720 723 725 There have been rare reports of N. gonorrhoeae with high-level ceftriaxone resistance in some countries (Japan, France, Spain).717 721 727 Although the overall prevalence of isolates with reduced susceptibility to cephalosporins remains low in the US, potential emergence of high-level cephalosporin resistance in N. gonorrhoeae is a major concern since available treatment options are limited if cephalosporins cannot be used.716 722

Susceptibility of N. gonorrhoeae in the US is being monitored by the CDC Gonococcal Isolate Surveillance Project (GISP).716 GISP data from 2000–2010 indicate that the percentage of US N. gonorrhoeae isolates with elevated ceftriaxone MICs (MICs 0.125 mcg/mL or greater) increased from 0.1 to 0.3% and the percentage with elevated cefixime MICs (MICs 0.25 mcg/mL or greater) increased from 0.2 to 1.4%.716 All 2009–2010 isolates with decreased susceptibility to cefixime were resistant to ciprofloxacin and tetracycline, but were susceptible to azithromycin.716 GISP data regarding N. gonorrhoeae isolates from men who have sex with men indicate that the percentage of isolates with elevated ceftriaxone MICs increased from 0% in 2006 to 1% in 2011 and the percentage of isolates with elevated cefixime MICs increased from 0.2% in 2006 to 3.8% in 2011.722 More recent GISP data indicate that the proportion of isolates in the US with decreased susceptibility to ceftriaxone or cefixime has remained low.344 During 2013, no isolates with decreased susceptibility to ceftriaxone or cefixime (i.e., MICs 0.5 mcg/mL or greater) were identified.344

Because of concerns related to reports of N. gonorrhoeae with reduced susceptibility to cephalosporins, CDC now recommends that IM ceftriaxone be used in conjunction with oral azithromycin for the treatment of uncomplicated gonorrhea (dual treatment).344 722 The theoretical basis of this recommendation is that combination therapy using 2 anti-infectives with different mechanisms of action may improve treatment efficacy and potentially delay emergence and spread of cephalosporin resistance.344 722 In addition, CDC no longer recommends use of cefixime or any other oral cephalosporin as first-line treatment for gonococcal infections.722 Cefixime should only be used as an alternative for the treatment of urogenital or rectal gonococcal infections when ceftriaxone is unavailable or cannot be used.344 722

CDC recommends that healthcare providers treating gonorrhea remain vigilant for treatment failures (evidenced by persistent symptoms or a positive follow-up test despite treatment) and report any occurrence of treatment failure to local or state health departments.716 722 If there is evidence of treatment failure, relevant clinical specimens should be cultured and in vitro susceptibility testing performed.344 722 In addition, an infectious disease specialist, an STD/HIV Prevention Training Center ([Web]), or the CDC (404-639-8659) should be consulted for treatment advice and the case should be reported to CDC through local or state health departments within 24 hours of diagnosis.344 722

Although a test-of-cure is not necessary for individuals with uncomplicated urogenital or rectal gonorrhea who are treated with a recommended or alternative regimen, those with pharyngeal gonorrhea who are treated with an alternative regimen should return 14 days after treatment for a test-of-cure.344 To minimize transmission, individuals treated for gonorrhea should be instructed to abstain from sexual activity for 7 days after treatment and until all sexual partners have been adequately treated.344 The patient's recent sexual partners (i.e., individuals having sexual contact with the infected patient within the 60 days preceding onset of symptoms or gonorrhea diagnosis) should be referred for evaluation, testing, and presumptive dual treatment.344

Uncomplicated Gonorrhea

For the treatment of uncomplicated cervical, urethral, or rectal gonorrhea in adults and adolescents, CDC states that a combination regimen that includes a single 250-mg dose of IM ceftriaxone and oral azithromycin (single 1-g dose) is the treatment of choice.344 Both drugs should be administered together on the same day, preferably simultaneously and under direct supervision.344

If necessary because ceftriaxone is not available or cannot be used and the patient has urogenital or rectal gonorrhea, CDC recommends an alternative combination regimen that includes a single 400-mg dose of oral cefixime and oral azithromycin (single 1-g dose).344

Disseminated Gonococcal Infections

CDC recommends that patients with disseminated gonococcal infections be hospitalized for initial treatment, especially when compliance may be a problem, when the diagnosis is uncertain, or when the patient has purulent synovial effusions or other complications.344 All patients with disseminated infections should be evaluated for clinical evidence of endocarditis and meningitis.344 Consultation with an infectious disease expert is advised.344

For treatment of disseminated gonococcal infections classified as arthritis and arthritis-dermatitis syndrome, CDC recommends that adults and adolescents receive a combination regimen of IM or IV ceftriaxone (1 g every 24 hours) and oral azithromycin (single 1-g dose);344 the recommended alternative is IV cefotaxime (1 g IV every 8 hours) and oral azithromycin (single 1-g dose).344 At 24–48 hours after substantial improvement, therapy can be switched to an oral anti-infective selected based on in vitro susceptibility testing.344 Total treatment duration for gonococcal arthritis and arthritis-dermatitis syndrome is at least 7 days.344

For treatment of disseminated gonococcal infections that involve meningitis and endocarditis, CDC recommends that adults and adolescents receive a combination regimen of IV ceftriaxone (1–2 g every 12–24 hours) and oral azithromycin (single 1-g dose).344 The parenteral regimen should be continued for 10–14 days for the treatment of gonococcal meningitis or for at least 4 weeks for the treatment of gonococcal endocarditis.344

Leptospirosis

Ceftriaxone and cefotaxime have been used in the treatment of severe leptospirosis caused by Leptospira.292 690 693 694 695 Leptospirosis is a spirochete infection that may range in severity from a self-limited systemic illness to a severe, life-threatening illness that includes jaundice, renal failure, hemorrhage, cardiac arrhythmias, pneumonitis, and hemodynamic collapse (Weil syndrome).292 691 693

Penicillin G generally has been considered the drug of choice for the treatment of moderate to severe leptospirosis.292 690 691 695 Other anti-infectives recommended for the treatment of severe leptospirosis include cephalosporins (ceftriaxone, cefotaxime), aminopenicillins (ampicillin, amoxicillin), tetracyclines (doxycycline, tetracycline), or macrolides (azithromycin).292 691 693 694 695

Lyme Disease

Oral cefuroxime axetil, IV ceftriaxone, and IV cefotaxime are used for the treatment of Lyme disease.292 329 348 470 471 472 473 474 476 480 481 482 483 484 485 486 487 488 489 492 493 514 666 First generation cephalosporins (e.g., cephalexin) are ineffective and should not be used for the treatment of Lyme disease.329 666 671 Although other second or third generation cephalosporins may be effective, the only cephalosporins currently recommended by IDSA, AAP, and other clinicians for the treatment of Lyme disease are cefuroxime axetil, ceftriaxone, and cefotaxime.292 329 470 471 472 473 474 476 480 481 482 483 484 485 486 487 488 489 492 493 514 666

Lyme disease (Lyme borreliosis) is a tick-born spirochetal disease.292 329 471 472 505 506 507 In the US, Lyme disease is caused by the spirochete Borrelia burgdorferi, which is transmitted by the bite of Ixodes scapularis or I. pacificus ticks.292 329 471 472 505 506 507 In addition to B. burgdorferi, I. scapularis may be simultaneously infected with and transmit Anaplasma phagocytophilum (causative agent of human granulocytotropic anaplasmosis [HGA, formerly known as human granulocytic ehrlichiosis]) and/or Babesia microti (causative agent of babesiosis).329 687 Because coinfection with A. phagocytophilum and/or B. microti can occur in patients with Lyme disease in geographic areas where these other pathogens are endemic, these diseases should be considered in the differential diagnosis of patients being evaluated for Lyme disease.329 687 Since cephalosporins are ineffective for the treatment of HGA and/or babesiosis, diagnosing such coinfections is critical to ensure that appropriate anti-infectives are used for treatment.329 687

Early Lyme Disease

Erythema migrans

IDSA, AAP, and other clinicians recommend oral doxycycline, oral amoxicillin, or oral cefuroxime axetil as first-line therapy for the treatment of the early localized or early disseminated Lyme disease associated with erythema migrans, in the absence of specific neurologic manifestations or advanced atrioventricular (AV) heart block.292 329 470 471 472 473 474 480 482 513 514 666 IDSA states that a 14-day regimen (range 14–21 days) of any of these oral anti-infectives (doxycycline, amoxicillin, cefuroxime axetil) may be used for initial treatment of early Lyme disease since all 3 drugs have been shown to be effective for the treatment of erythema migrans and associated symptoms in prospective clinical studies.329 Doxycycline offers the advantage of also being effective for the treatment of HGA coinfection (but not babesiosis coinfection).292 329

Although IV ceftriaxone is effective for early Lyme disease manifested as erythema migrans, it is not superior to the recommended oral anti-infectives and is more likely to cause serious adverse effects; therefore, IV ceftriaxone is not usually recommended for the treatment of early Lyme disease in the absence of neurologic involvement or advanced AV heart block.329 666

Transplacental transmission of B. burgdorferi appears to occur rarely, if at all,666 and epidemiologic studies in pregnant women have not documented an association between exposure to Lyme disease prior to conception or during pregnancy and subsequent fetal death, congenital malformations, or prematurity.480 487 IDSA, AAP, and other clinicians state that pregnant or nursing women need not be treated differently than other patients with Lyme disease, except that they should not receive doxycycline.292 329 480 666

Early Neurologic Lyme Disease

Although oral anti-infectives (e.g., doxycycline, amoxicillin, cefuroxime axetil) generally are effective for the treatment of the early Lyme disease associated with erythema migrans in the absence of specific neurologic manifestations, parenteral anti-infectives are recommended for the treatment of early Lyme disease when there are acute neurologic manifestations such as meningitis or radiculopathy.292 329 470 471 472 498 499 501 502 513 515 516 666 Radiculopathy, cranial neuropathy, and mononeuropathy multiplex may be manifestations of acute peripheral nervous system involvement.329 In the US, cranial neuropathy is the most common manifestation of early neurologic Lyme disease; seventh cranial nerve palsy is the most common of the cranial neuropathies and bilateral involvement may occur.329 Cranial nerve palsies in patients with Lyme disease frequently are associated with lymphocytic CSF pleocytosis, with or without symptoms of meningitis.329 Although anti-infectives may not hasten resolution of seventh cranial nerve palsy associated with B. burgdorferi infection, anti-infectives should be given to prevent further sequelae.329

IDSA and other clinicians recommend a 14-day regimen (range: 10–28 days) of IV ceftriaxone as first-line therapy for the treatment of acute neurologic Lyme disease manifested by meningitis or radiculopathy; IV cefotaxime and IV penicillin G sodium are the preferred alternatives.292 329 666 It has been suggested that IV ceftriaxone may be preferable to IV penicillin G for serious manifestations of early disseminated or late Lyme disease (i.e., those involving major organs) based on ceftriaxone’s greater in vitro and in vivo activity against B. burgdorferi, excellent CSF penetration, and prolonged serum concentrations achievable with once-daily administration of the drug.329 471 472 504 509 510 511 512 Although IV cefotaxime appears to be as effective as IV ceftriaxone or IV penicillin G sodium for the treatment of acute neurologic Lyme disease and does not cause the biliary complications reported with ceftriaxone, ceftriaxone has the advantage of once-daily dosing.329 470 514 516 666 686 In patients with acute neurologic manifestations who are intolerant of cephalosporins and penicillin, there is some evidence that oral doxycycline may be an adequate alternative that can be considered for use in adults and children 8 years of age or older; because doxycycline is well absorbed orally, IV doxycycline should only be needed rarely.329

Some clinicians suggest that a 14-day regimen (range: 14–21 days) of oral anti-infectives (amoxicillin, doxycycline, cefuroxime axetil) may be used in patients with cranial nerve palsy without clinical evidence of meningitis (i.e., those with normal CSF examinations or those for whom CSF examination is deemed unnecessary because there are no clinical signs of meningitis); however, a 14-day parenteral regimen (range: 10–28 days) is indicated when there is both clinical and laboratory evidence of CNS involvement and meningitis.292 329 Although there is some experience using oral anti-infectives in patients with seventh cranial nerve palsy, it is unclear whether an oral regimen would be as effective for patients with other cranial neuropathies.329

Lyme Carditis

IDSA states that patients with AV heart block and/or myopericarditis associated with early Lyme disease may be treated with a 14-day regimen (range: 14–21 days) of oral or parenteral anti-infectives.329 Although there is no evidence to date to suggest that a parenteral regimen is more effective than an oral regimen for the treatment of Lyme carditis, a parenteral regimen usually is recommended for initial treatment of hospitalized patients; an oral regimen can be used to complete therapy and for the treatment of outpatients.292 329 470 666 When a parenteral regimen is used, IV ceftriaxone or, alternatively, IV cefotaxime or IV penicillin G sodium is recommended.292 329 470 666 When an oral regimen is used, oral amoxicillin, oral doxycycline, or oral cefuroxime axetil is recommended.292 329 470 666

Because of the potential for life-threatening complications, hospitalization and continuous monitoring is advisable for patients who are symptomatic (syncope, dyspnea, chest pain) and also is recommended for those with second- or third-degree AV block or first-degree heart block when the PR interval is prolonged to 0.3 seconds or longer.329 666 Patients with advanced heart block may require a temporary pacemaker and consultation with a cardiologist is recommended.329 666

Borrelial Lymphocytoma

Although experience is limited, IDSA states that available data indicate that borrelial lymphocytoma may be treated with a 14-day regimen (range: 14–21 days) of oral doxycycline, oral amoxicillin, or oral cefuroxime axetil in the dosages used for the treatment of erythema migrans.329

Late Lyme Disease

Lyme Arthritis

Patients with uncomplicated Lyme arthritis without clinical evidence of neurologic disease generally can be treated with a 28-day regimen of oral doxycycline, oral amoxicillin, or oral cefuroxime axetil.292 329 470 480 666 669 However, patients with Lyme arthritis and concomitant neurologic disease should receive a 14-day parenteral regimen (range: 14–28 days) of IV ceftriaxone or, alternatively, IV cefotaxime or IV penicillin G sodium.292 329 470 666 While oral regimens are easier to administer, associated with fewer serious adverse effects, and less expensive than IV regimens, some patients with Lyme arthritis treated with oral anti-infectives have subsequently developed overt neuroborreliosis, which may require IV therapy for successful resolution.329 Therefore, additional study is needed to fully evaluate the comparative safety and efficacy of oral versus IV anti-infectives for the treatment of Lyme arthritis.329

In patients who have persistent or recurrent joint swelling after a recommended oral regimen, IDSA and other clinicians recommend retreatment with the oral regimen or a switch to a parenteral regimen.329 470 480 666 Some clinicians prefer retreatment with an oral regimen for patients whose arthritis substantively improved but did not completely resolve; these clinicians reserve parenteral regimens for those patients whose arthritis failed to improve or worsened.329 It has been suggested that clinicians should consider allowing several months for joint inflammation to resolve after initial treatment before an additional course of anti-infectives is given.329

Late Neurologic Lyme Disease

IDSA and other clinicians recommend that patients with late neurologic Lyme disease affecting the CNS or peripheral nervous system (e.g., encephalopathy, neuropathy) receive a 14-day regimen (range: 14–28 days) of IV ceftriaxone or, alternatively, IV cefotaxime or IV penicillin G sodium.329 Response to anti-infective treatment usually is slow and may be incomplete in patients with late neurologic Lyme disease.329 IDSA states that retreatment is not recommended unless relapse is shown by reliable objective measures.329

Acrodermatitis Chronica Atrophicans

IDSA states that available data indicate that acrodermatitis chronica atrophicans may be treated with a 21-day regimen (range: 14–28 days) of oral doxycycline, oral amoxicillin, or oral cefuroxime axetil in the dosages used for the treatment of erythema migrans.329 It is unclear whether a parenteral regimen would be more effective than an oral regimen.329

Nocardiosis

Cephalosporins (ceftriaxone, cefotaxime, cefoxitin, cefuroxime) have been used for the treatment of nocardiosis caused by Nocardia.292 697 698 699 700 701 702

Co-trimoxazole (fixed combination of sulfamethoxazole and trimethoprim) usually is considered the drug of choice for the treatment of nocardiosis.292 Other drugs that have been used alone or in combination regimens for the treatment of nocardiosis include a sulfonamide alone (sulfadiazine, sulfamethoxazole [not commercially available as a single-entity preparation in the US]), amikacin, tetracyclines, cephalosporins (ceftriaxone, cefotaxime, cefuroxime), cefoxitin, carbapenems (imipenem or meropenem), fixed combination of amoxicillin and clavulanate, clarithromycin, cycloserine, or linezolid.292 697 698 700 701 702 710

Anti-infective agents for the treatment of invasive nocardiosis or for the treatment of nocardiosis in patients unable to tolerate sulfonamides should be selected based on results of in vitro susceptibility testing.292 If nocardiosis involves the CNS or if the infection is disseminated or overwhelming, some clinicians suggest that amikacin and ceftriaxone should be included in the treatment regimen during the first 4–12 weeks of therapy or until there is clinical improvement.292 A regimen of amikacin and ceftriaxone has been effective for the treatment of disseminated N. asteroides infection complicated by cerebral abscess.698

Pelvic Inflammatory Disease

Several parenteral cephalosporins (cefotaxime332 344 , ceftriaxone343 344 ) and closely related cephamycins (cefotetan313 344 , cefoxitin344 ) have been used in the treatment of acute pelvic inflammatory disease (PID); these drugs are inactive against C. trachomatis and should not be used alone in the treatment of PID.344

PID is an acute or chronic inflammatory disorder in the upper female genital tract and can include any combination of endometritis, salpingitis, tubo-ovarian abscess, and pelvic peritonitis.344 417 PID generally is a polymicrobial infection frequently caused by sexually transmitted organisms, especially N. gonorrhoeae and/or Chlamydia trachomatis; however, organisms that can be part of the normal vaginal flora (e.g., anaerobic bacteria, Gardnerella vaginalis, H. influenzae, enteric gram-negative bacilli, S. agalactiae) also have been associated with PID.344 In addition, cytomegalovirus or mycoplasma (e.g., Mycoplasma hominis, M. genitalium, Ureaplasma urealyticum) may be involved in some cases.344 417 418 PID is treated with an empiric regimen that provides broad-spectrum coverage.344 417 418 The regimen should be effective against N. gonorrhoeae and C. trachomatis and also probably should be effective against anaerobes, gram-negative facultative bacteria, and streptococci.344 417 419 The optimum empiric regimen for the treatment of PID has not been identified.344 A wide variety of parenteral and oral regimens have been shown to achieve clinical and microbiologic cure in randomized studies with short-term follow-up;344 418 419 however, only limited data are available to date regarding elimination of infection in the endometrium and fallopian tubes or intermediate or long-term outcomes, including the impact of these regimens on the incidence of long-term sequelae of PID (e.g., tubal infertility, ectopic pregnancy, pain).344 419

Parenteral Regimens for PID

When a parenteral regimen is indicated for the treatment of PID, CDC generally recommends a 2-drug regimen of cefoxitin (2 g IV every 6 hours) or cefotetan (2 g IV every 12 hours) given in conjunction with doxycycline (100 mg IV or orally every 12 hours) or a 2-drug regimen of clindamycin (900 mg IV every 8 hours) and gentamicin (usually a 2-mg/kg IV or IM loading dose followed by 1.5 mg/kg every 8 hours).344 While certain parenteral cephalosporins (e.g., cefotaxime, ceftriaxone) also have been used for the treatment of PID,344 417 418 420 CDC states that only limited data are available regarding use of these cephalosporins in patients with PID and they are less active than cefotetan or cefoxitin against anaerobic bacteria.344 CDC states that only limited data are available to support the use of other parenteral regimens for the treatment of acute PID, although a regimen of IV ampicillin and sulbactam given with oral or IV doxycycline provides broad-spectrum coverage and is effective against C. trachomatis, N. gonorrhoeae, and anaerobes in women with tubo-ovarian abscess.344

Oral Regimens for PID

Data are not available regarding use of oral cephalosporins for the treatment of PID.344 When acute PID that is mild to moderately severe is treated with an oral regimen, CDC recommends a regimen that consists of a single dose of IM ceftriaxone, IM cefoxitin (with oral probenecid), or IM cefotaxime given in conjunction with a 14-day regimen of oral doxycycline (with or without oral metronidazole).344 The optimal parenteral cephalosporin for this regimen is unclear; although cefoxitin has better anaerobic coverage, ceftriaxone has better coverage against N. gonorrhoeae.344

For additional information regarding treatment of PID, the current CDC sexually transmitted diseases treatment guidelines available at [Web] should be consulted.344

Syphilis

Ceftriaxone has some activity against Treponema pallidum173 and there is some limited evidence that the drug may be effective for the treatment of syphilis.216 228 260 344 Penicillin G is the drug of choice for the treatment of all stages of syphilis and data to support the use of penicillin alternatives are limited.344

CDC states that, based on limited clinical studies, biologic plausibility, and pharmacologic properties, ceftriaxone may be an effective alternative for the treatment of primary or secondary syphilis or neurosyphilis in penicillin-allergic patients.344 However, optimal dosage and duration of ceftriaxone therapy for these infections have not been defined and the possibility of cross-allergenicity with penicillin should be considered.344 Although ceftriaxone might be effective for the treatment of latent syphilis in penicillin-allergic patients, CDC states that the only acceptable alternatives to penicillin for the treatment of these infections are doxycycline or tetracycline.344 Decisions regarding the treatment of syphilis in penicillin-allergic patients should be made in consultation with a specialist.344

For the treatment of infants with clinical evidence of congenital syphilis, CDC states that use of ceftriaxone can be considered if there is a penicillin shortage and penicillin G sodium and penicillin G procaine are unavailable.344 However, because studies that strongly support ceftriaxone for the treatment of congenital syphilis have not been conducted, the drug should be used with careful clinical and serologic follow-up and in consultation with a specialist in the treatment of infants with congenital syphilis.344

For additional information regarding treatment of syphilis, the current CDC sexually transmitted diseases treatment guidelines available at [Web] should be consulted.344

Empiric Therapy in Febrile Neutropenic Patients

Cefepime has been used alone or in conjunction with other anti-infectives for empiric anti-infective therapy of presumed bacterial infections in febrile neutropenic adults or pediatric patients.314 457 459 466 467 468 469 Ceftazidime454 455 457 459 468 and ceftriaxone454 455 also have been used for empiric anti-infective therapy in febrile neutropenic patients, but are not generally recommended.453 457 463 Ceftazidime is no longer considered a reliable drug for empiric monotherapy in febrile neutropenic patients because of poor activity against gram-positive bacteria and decreasing potency against gram-negative bacteria;457 ceftriaxone monotherapy may not provide adequate coverage against some potential pathogens (e.g., Ps. aeruginosa).453 463

Successful treatment of infections in granulocytopenic patients requires prompt initiation of empiric anti-infective therapy (even when fever is the only sign or symptom of infection) and appropriate modification of the initial regimen if the duration of fever and neutropenia is protracted, if a specific site of infection is identified, or if organisms resistant to the initial regimen are present.457 458 459 460 462 The initial empiric regimen should be chosen based on the underlying disease and other host factors that may affect the degree of risk, on local epidemiologic data regarding usual pathogens in these patients and in vitro susceptibility of bacterial isolates recovered from other patients in the same health-care facility, and the individual patient's pattern of colonization and resistance.457 458 460 461 462 The fact that gram-positive bacteria have become a predominant pathogen in febrile neutropenic patients should be considered when selecting an empiric anti-infective regimen.457 458 460 However, although gram-positive bacteria reportedly account for about 60% and gram-negative bacteria account for about 35% of microbiologically documented infections, gram-negative infections are associated with greater mortality.457

No empiric regimen has been identified that would be appropriate for initial treatment of all febrile neutropenic patients.457 458 460 462 IDSA recommends use of a parenteral empiric regimen in most febrile neutropenic patients; use of an oral regimen (e.g., oral ciprofloxacin and oral amoxicillin and clavulanate potassium) should be considered only for selected adults at low risk for complications who have adequate and stable renal and hepatic function, an expected duration of neutropenia less than 7 days, and no active medical comorbidities.457 All other patients are considered high risk and should receive an initial IV empiric regimen consisting of an antipseudomonal β-lactam (e.g., cefepime, imipenem, meropenem, fixed-combination of piperacillin and tazobactam).457 Other anti-infectives (e.g., aminoglycosides, fluoroquinolones, and/or vancomycin) may be added to the regimen for the management of complications (e.g., pneumonia) or when antimicrobial resistance is suspected or proven.457 Vancomycin and other anti-infectives active against aerobic gram-positive cocci are not usually included in the initial empiric regimen except in certain clinical scenarios, including suspected catheter-related infections, skin and soft tissue infections, pneumonia, or hemodynamic instability.457

Regardless of the initial regimen selected, patients should be reassessed daily and the anti-infective regimen altered (if indicated) based on the presence or absence of fever, identification of the causative organism, and the clinical condition of the patient; anti-infectives active against gram-positive organisms may be discontinued after 2 days if there is no evidence of such infections.457

Published protocols for the treatment of infections in febrile neutropenic patients should be consulted for specific recommendations regarding selection of the initial empiric anti-infective regimen, when to change the initial regimen, possible subsequent regimens, and duration of anti-infective therapy in these patients.457 In addition, consultation with an infectious disease expert knowledgeable about infections in immunocompromised patients is advised.457

Perioperative Prophylaxis

Cefazolin, cefotaxime, ceftriaxone, and cefuroxime have been used perioperatively to reduce the incidence of infections in patients undergoing certain contaminated or potentially contaminated surgical procedures or undergoing certain clean surgical procedures where the development of infection at the surgical site would have severe consequences.310 332 340 343 347 354 355 356 357 358 360 374 629 639 640 641 643 644 Perioperative prophylaxis with an appropriate anti-infective agent can decrease the incidence of infection, particularly surgical site infections, after some procedures.360

Because cefazolin has a narrow spectrum of activity that covers the most likely surgical site pathogens, has a moderately long serum half-life, and has been shown to be effective, it is considered by many clinicians to be the drug of choice for perioperative prophylaxis for a wide variety of procedures.360 Cefazolin is considered a drug of choice for perioperative prophylaxis in patients undergoing certain cardiac surgery (e.g., coronary artery bypass, placement of pacemaker or other cardiac device), noncardiac thoracic surgery (e.g., lobectomy), vascular surgery (arterial surgery involving the abdominal aorta, a prosthesis, or a groin incision or lower extremity amputation for ischemia), head and neck surgery involving incisions through oral or pharyngeal mucosa, neurosurgery (e.g., craniotomy, spinal surgery), orthopedic surgery (e.g., total joint replacement, surgical repair of closed fractures, internal fixation of compound or open fractures), GI surgery (gastroduodenal, esophageal, biliary tract, colorectal, appendectomy, bariatric), genitourinary surgery (e.g., open or laparoscopic surgery including percutaneous renal surgery, procedures with entry into the urinary tract or implantation of a prosthesis), and gynecologic and obstetric surgery (cesarean section or vaginal, abdominal, or laparoscopic hysterectomy).360 374 Cefazolin also is recommended as a drug of choice for perioperative prophylaxis in patients undergoing heart, lung, heart-lung, pancreas, and pancreas-kidney transplantation.374

Cefuroxime is considered a drug of choice for cardiac procedures (e.g., coronary artery bypass, placement of pacemaker or other cardiac device).360 374 Cefuroxime also is considered a drug of choice for perioperative prophylaxis in patients undergoing clean head and neck surgery involving placement of prosthesis (excluding tympanostomy) and is a drug of choice when used in conjunction with metronidazole for perioperative prophylaxis in patients undergoing clean-contaminated cancer surgery of the head and neck or other clean-contaminated head and neck procedures (excluding tonsillectomy and functional endoscopic sinus procedures).374

Cefotaxime used in conjunction with ampicillin is considered a regimen of choice for perioperative prophylaxis in patients undergoing liver transplantation.374

Ceftriaxone is recommended as one of several options for perioperative prophylaxis in patients undergoing biliary tract procedures, but should not be used in patients undergoing cholecystectomy for noninfected biliary conditions.374

A first or second generation cephalosporin (cefazolin, cefuroxime) generally is preferred when a cephalosporin is used for perioperative prophylaxis.360 374 Third generation cephalosporins (cefotaxime, ceftriaxone, ceftazidime) and fourth generation cephalosporins (cefepime) are not usually recommended for perioperative prophylaxis because they are expensive, some are less active than first or second generation cephalosporins against staphylococci, they have spectrums of activity wider than necessary for organisms encountered in elective surgery, and their use for prophylaxis may promote emergence of resistant organisms.360 374

If cefazolin is used for perioperative prophylaxis in patients undergoing certain GI procedures (e.g., colorectal surgery, appendectomy) that might involve exposure to B. fragilis or other anaerobic bowel bacteria or in patients undergoing head and neck surgery involving incisions through oral or pharyngeal mucosa, it should be used in conjunction with metronidazole to provide anaerobic coverage.360 374

Published guidelines and protocols for perioperative prophylaxis should be consulted for additional information regarding specific procedures.360 374

Cardiac Surgery

Perioperative prophylaxis can decrease the incidence of infection after cardiac surgery.360 639 For cardiac procedures lasting longer than 400 minutes, the risk of postoperative infection is decreased if intraoperative anti-infective doses are given in addition to a preoperative dose.360 Regarding the use of postoperative doses in patients undergoing cardiac surgery, there are data to support use of a single preoperative dose or prophylaxis continued for 24 hours postoperatively; however, there is no evidence of benefit beyond 48 hours and no evidence to support continuing prophylaxis after wound closure or until all indwelling drains and intravascular catheters are removed.360 374

Results of a pooled analysis of 7 placebo-controlled studies indicate that perioperative prophylaxis reduces the incidence of infection after permanent pacemaker implantation.639 In patients receiving implantation of permanent pacemakers and cardioverter-defibrillators, studies indicate that perioperative prophylaxis reduces the incidence of wound infection, inflammation, and skin erosion.360

For perioperative prophylaxis in patients undergoing cardiac surgery, many clinicians recommend IV cefazolin or IV cefuroxime.360 374 Although routine use of vancomycin for perioperative prophylaxis is not recommended, IV vancomycin is considered an alternative for perioperative prophylaxis in cardiac surgery patients at institutions where methicillin-resistant S. aureus (MRSA; also known as oxacillin-resistant S. aureus or ORSA) and methicillin-resistant S. epidermidis frequently cause postoperative wound infection or in patients who were previously colonized with MRSA or are allergic to penicillins or cephalosporins.360

Noncardiac Thoracic Surgery

Although studies evaluating use of perioperative prophylaxis in patients undergoing pulmonary surgery are sparse, such prophylaxis is routinely used.360 In one randomized, double-blind, placebo-controlled study in patients undergoing noncardiac thoracic surgery, a single preoperative dose of cefazolin decreased the incidence of postoperative surgical site infection, but did not affect the incidence of postoperative empyema or nosocomial pneumonia.641 Although there is evidence from a placebo-controlled study that multiple doses of a cephalosporin (cefazolin) can prevent infection after closed-tube thoracostomy for chest trauma,640 some clinicians suggest that insertion of chest tubes for other indications, such as spontaneous pneumothorax, does not require prophylaxis.360

For perioperative prophylaxis in patients undergoing noncardiac thoracic surgery, many clinicians recommend IV cefazolin or IV ampicillin and sulbactam.360 374 Although routine use of vancomycin for perioperative prophylaxis is not recommended, IV vancomycin may be considered an alternative for perioperative prophylaxis in patients undergoing noncardiac thoracic surgery at institutions where MRSA and methicillin-resistant S. epidermidis frequently cause postoperative wound infection or in patients who were previously colonized with MRSA or are allergic to penicillins or cephalosporins.360 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin or vancomycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360 374

GI Surgery

Esophageal and Gastroduodenal Surgery

Perioperative prophylaxis is not usually indicated for patients undergoing routine gastroesophageal endoscopy, but is recommended for high-risk patients undergoing esophageal or gastroduodenal procedures.360

Although there are no controlled studies evaluating efficacy, perioperative prophylaxis is used routinely in patients undergoing bariatric surgery, including adjustable gastric banding, vertical banded gastroplasty, Roux-en-y bypass, and biliopancreatic diversion.360

For perioperative prophylaxis in high-risk patients undergoing esophageal or gastroduodenal surgery (e.g., those who are morbidly obese or have GI obstruction, decreased gastric acidity, decreased GI motility, gastric bleeding, malignancy or perforation, or immunosuppression), many clinicians recommend IV cefazolin as the drug of choice.360 374 Dosage may need to be increased in morbidly obese patients.360 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin or vancomycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360 374

Biliary Tract Surgery

Perioperative prophylaxis is recommended for patients undergoing biliary tract surgery who are at high risk of infection (e.g., those older than 70 years of age or those with acute cholecystitis, a nonfunctioning gallbladder, obstructive jaundice, or common duct stones).360 In high-risk patients undergoing endoscopic retrograde cholangiopancreatography (ERCP), perioperative prophylaxis is recommended only if complete biliary drainage is unlikely to be achieved.360 Perioperative prophylaxis is not considered necessary for low-risk patients undergoing elective laparoscopic cholecystectomy360 374 635 since there is no evidence that such prophylaxis provides any benefit in these patients.635

For perioperative prophylaxis in high-risk patients undergoing biliary tract surgery (e.g., those older than 70 years of age or those with acute cholecystitis, nonfunctioning gallbladder, obstructive jaundice, or common duct stones), many clinicians recommend IV cefazolin as the drug of choice.360 374 Alternatives include other cephalosporins (cefotetan, cefoxitin, ceftriaxone) or ampicillin and sulbactam.374 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin or vancomycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360 374

Colorectal Surgery

There is evidence that perioperative prophylaxis can decrease the incidence of infection after colorectal surgery, and such prophylaxis usually is recommended.360

For perioperative prophylaxis in patients undergoing colorectal surgery, many clinicians recommend a parenteral regimen of IV cefoxitin or IV cefotetan, IV cefazolin (or IV ceftriaxone) used in conjunction with IV metronidazole, or IV ampicillin and sulbactam.360 374 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360 374

Alternatively, an oral prophylaxis regimen of oral neomycin with either oral erythromycin or oral metronidazole can be used.360 Many clinicians use both an oral regimen and a parenteral regimen for perioperative prophylaxis in patients undergoing colorectal surgery,360 374 637 and there is some evidence that use of an oral regimen (with mechanical bowel preparation) in conjunction with a parenteral regimen is more effective than use of a parenteral regimen alone.360

In a randomized, prospective study in patients undergoing elective colorectal surgery, the overall incidence of intra-abdominal septic complications in those who received mechanical bowel preparation and an oral regimen (erythromycin and neomycin) alone was similar to that in those who received both the oral regimen and a parenteral regimen (cefoxitin); however, the incidence of abdominal wound infection was higher in those who received the oral regimen alone (14.6%) than in those who received the combined oral and parenteral regimen (5%).638

Appendectomy

There is evidence that perioperative prophylaxis can reduce the incidence of infection after surgery for acute appendicitis.360

For perioperative prophylaxis in patients undergoing nonperforated appendectomy, many clinicians recommend IV cefoxitin or IV cefotetan or IV cefazolin used in conjunction with IV metronidazole.360 374 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin or vancomycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360 374

If perforation has occurred, anti-infectives are continued postoperatively for about 5 days.360

Genitourinary Surgery

Cystoscopy

Perioperative prophylaxis generally is not recommended in patients with sterile urine undergoing cystoscopy without manipulation (dilation, biopsy, fulguration, resection or ureteral instrumentation).360 374 When cystoscopy with manipulation is planned, urine cultures are positive or unavailable, or an indwelling urinary catheter is present, many clinicians recommend that patients receive treatment with an appropriate anti-infective before surgery to sterilize the urine or receive a single preoperative dose of an anti-infective selected based on the most likely infecting organisms.360

If perioperative prophylaxis is indicated in individuals undergoing cystoscopy who are at high risk (urine culture positive or unavailable, preoperative urinary catheter, transrectal prostatic biopsy, placement of prosthetic material) or in individuals undergoing cystoscopy with manipulation or upper urinary tract instrumentation (shockwave lithotripsy, ureteroscopy), many clinicians recommend oral or IV ciprofloxacin, oral co-trimoxazole, or IV cefazolin.360 374

Open or Laparoscopic Surgery

There is evidence that perioperative prophylaxis decreases the incidence of postoperative bacteriuria and septicemia in patients with sterile urine undergoing transurethral prostatectomy and transrectal prostatic biopsies.360 Prophylaxis also is used for patients undergoing ureteroscopy, shock wave lithotripsy, percutaneous renal surgery, or open laparoscopic procedures or undergoing placement of a urologic prosthesis (penile implant, artificial sphincter, synthetic pubovaginal sling, bone anchors for pelvic floor reconstruction).360

For perioperative prophylaxis in individuals undergoing open or laparoscopic surgery (including percutaneous renal surgery, procedures with entry into the urinary tract, or procedures involving implantation of a prosthesis), many clinicians recommend IV cefazolin.360 In patients allergic to penicillins and cephalosporins, a reasonable alternative is clindamycin used in conjunction with gentamicin, ciprofloxacin, levofloxacin, or aztreonam.360

Gynecologic and Obstetric Surgery

Perioperative prophylaxis decreases the incidence of infection after vaginal and abdominal hysterectomy.360 In addition, there is evidence that perioperative prophylaxis can prevent infection after elective and nonelective cesarean section, including emergency cesarean section in high-risk situations (e.g., active labor, premature rupture of membranes), and after elective abortions, including second trimester abortions or first trimester abortion in high-risk women.360 374 623 A pooled analysis of results of randomized, placebo-controlled studies in women who underwent therapeutic abortion before 16 weeks’ gestation indicates that perioperative prophylaxis reduces the overall risk of postabortal infection by 42% compared with placebo.623

Many clinicians suggest that the preferred agents for perioperative prophylaxis in women undergoing vaginal, abdominal, or laparoscopic hysterectomy are IV cefoxitin, IV cefotetan, IV cefazolin, or IV ampicillin and sulbactam.360 374 IV cefazolin generally is the preferred agent for prophylaxis in women undergoing cesarean section;360 374 oral doxycycline is recommended for prophylaxis in those undergoing abortion.360

Head and Neck Surgery

There is evidence that perioperative prophylaxis decreases the incidence of surgical site infection after oncologic head and neck operations involving incisions through the oral or pharyngeal mucosa.360 Perioperative prophylaxis has not been shown to be beneficial in patients undergoing tonsillectomy, nasal septoplasty, or functional endoscopic sinus procedures.360 374 Perioperative prophylaxis is not indicated for clean head and neck surgery.374

For perioperative prophylaxis in patients undergoing head or neck surgery involving incisions through oral or pharyngeal mucosa, many clinicians recommend IV clindamycin, IV cefazolin used in conjunction with IV metronidazole, or IV ampicillin and sulbactam.360 Other clinicians recommend cefazolin or cefuroxime for perioperative prophylaxis in patients undergoing clean head and neck surgery involving placement of prosthesis (excluding tympanostomy).374 For patients undergoing clean-contaminated cancer surgery of the head and neck or other clean-contaminated head and neck procedures (excluding tonsillectomy and functional endoscopic sinus procedures), these experts recommend cefazolin used in conjunction with metronidazole, cefuroxime used in conjunction with metronidazole, or ampicillin and sulbactam.374

Neurosurgery

There is evidence that perioperative use of an anti-infective agent active against staphylococci can decrease the incidence of infection after craniotomy.360 Although some studies in patients undergoing implantation of permanent CSF shunts showed lower infection rates in those receiving perioperative prophylaxis, the benefit of such prophylaxis in patients undergoing ventriculostomy placement remains uncertain.360

Although the rate of postoperative infections is low in patients undergoing spinal surgery involving conventional lumbar discectomy, many clinicians use perioperative prophylaxis in such patients because of the serious consequences of a surgical site infection.360 The risk of infection is higher after prolonged spinal surgery or spinal procedures involving fusion or insertion of foreign material, and perioperative prophylaxis generally is used in these patients.360

For perioperative prophylaxis in patients undergoing neurosurgery, many clinicians recommend IV cefazolin.360 374 Although routine use of vancomycin for perioperative prophylaxis is not recommended, IV vancomycin may be considered an alternative for perioperative prophylaxis in patients undergoing neurosurgery at institutions where MRSA and methicillin-resistant S. epidermidis frequently cause postoperative wound infection or in patients who were previously colonized with MRSA or are allergic to penicillins or cephalosporins.360

Ophthalmic Surgery

Data are limited regarding the effectiveness of anti-infective prophylaxis in patients undergoing ophthalmic surgery; however, many clinicians use a topical or subconjunctival anti-infective for prophylaxis in patients undergoing such surgery.360 Some clinicians state that there is no evidence that perioperative prophylaxis is needed for procedures that do not invade the globe.360 While there is no consensus regarding the most effective drug, route, or duration of perioperative prophylaxis in patients undergoing ophthalmic surgery, many clinicians recommend use of a topical aminoglycoside (gentamicin, tobramycin), topical fluoroquinolone (ciprofloxacin, gatifloxacin, levofloxacin, moxifloxacin, ofloxacin), or a topical preparation containing neomycin, polymyxin b sulfate, and gramicidin; some clinicians also recommend subconjunctival injection of cefazolin or intracameral cefazolin or cefuroxime.360 374

Orthopedic Surgery

There is evidence that perioperative prophylaxis with an anti-infective agent active against staphylococci can decrease the incidence of both early and delayed infection in patients undergoing joint replacement or surgical repair of closed fractures.360 Perioperative prophylaxis can decrease the rate of infection in patients with hip and other closed fractures undergoing procedures that involve internal fixation with nails, plates, screws, or wires and in patients with compound or open fractures.360 However, additional study is needed to determine whether single-dose regimens or regimens continued for 24 hours are most effective.360 A retrospective review concluded that anti-infective prophylaxis is not indicated for patients undergoing arthroscopic surgery procedures.360

For perioperative prophylaxis in patients undergoing orthopedic surgery, many clinicians recommend IV cefazolin.360 374 Although routine use of vancomycin for perioperative prophylaxis is not recommended, IV vancomycin may be considered an alternative for perioperative prophylaxis in patients undergoing orthopedic surgery at institutions where MRSA and methicillin-resistant S. epidermidis frequently cause postoperative wound infection or in patients who were previously colonized with MRSA or are allergic to penicillins or cephalosporins.360

Vascular Surgery

There is evidence that perioperative prophylaxis with a cephalosporin can decrease the incidence of postoperative surgical site infection after arterial reconstructive surgery on the abdominal aorta, vascular operations on the leg that include a groin incision, or amputation of a lower extremity for ischemia.360 Many clinicians also recommend perioperative prophylaxis in patients undergoing implantation of any vascular prosthetic material (e.g., grafts for vascular access in hemodialysis); however, such prophylaxis generally is not indicated for carotid endarterectomy or brachial artery repair without prosthetic material.360 Although perioperative prophylaxis is not routinely recommended for endovascular stenting, prophylaxis may be justified in patients with certain risk factors (e.g., repeat intervention within 7 days, prolonged indwelling arterial sheath, procedure duration longer than 2 hours, presence of other infected implants, immunosuppression).360

For perioperative prophylaxis in patients undergoing vascular surgery (e.g., arterial surgery involving a prosthesis, the abdominal aorta, or a groin incision; lower extremity amputation for ischemia), many clinicians recommend IV cefazolin.360 374 Although routine use of vancomycin for perioperative prophylaxis is not recommended, IV vancomycin may be considered an alternative for perioperative prophylaxis in patients undergoing vascular surgery at institutions where MRSA and methicillin-resistant S. epidermidis frequently cause postoperative wound infection or in patients who were previously colonized with MRSA or are allergic to penicillins or cephalosporins.360

Timing and Number of Doses

When perioperative prophylaxis is indicated in patients undergoing surgery, administration of an appropriate anti-infective should be timed to ensure that bactericidal concentrations of the drug are established in serum and tissues by the time the initial surgical incision is made;360 374 therapeutic concentrations of the drug should then be maintained in serum and tissues for the duration of the procedure.374

An IV dose of the appropriate anti-infective should be given within 60 minutes before the initial incision.360 However, if vancomycin or a fluoroquinolone is used, the anti-infective infusion should be started within 1–2 hours before the initial incision to minimize the risk of an adverse effect occurring around the time of anesthesia induction and to ensure adequate tissue levels of the drug at the time of the initial incision.360 374

Single-dose perioperative prophylaxis may be sufficient.374 However, if surgery is prolonged (more than 3–4 hours), major blood loss occurs, or other factors are present that shorten the half-life of the drug (e.g., extensive burns), additional doses should be administered during the procedure to ensure adequate serum and tissue concentrations of the anti-infective throughout the procedure.360 374 Intraoperative doses may not be warranted if there are factors that prolong the half-life of the drug (e.g., renal impairment).374 For prolonged procedures in patients with normal renal function, some clinicians suggest that intraoperative doses be administered during the procedure at intervals that correspond to 1–2 times the half-life of the drug.360 If intraoperative doses are necessary, the redosing interval should be measured from the time of administration of the preoperative dose, not from the beginning of the procedure.374

For most procedures, the duration of prophylaxis should be less than 24 hours.360 374 There is no evidence to support continuing prophylaxis after wound closure or until all indwelling drains and intravascular catheters are removed.360 374

Prevention of Perinatal Group B Streptococcal Disease

In certain penicillin-allergic women, cefazolin is used as an alternative to penicillin G or ampicillin for prevention of perinatal group B streptococcal (GBS) disease.359

Pregnant women who are colonized with GBS in the genital or rectal areas can transmit GBS infection to their infants during labor and delivery resulting in invasive neonatal infection that can be associated with substantial morbidity and mortality.359 Intrapartum anti-infective prophylaxis for prevention of early-onset neonatal GBS disease is administered selectively to women at high risk for transmitting GBS infection to their neonates.359 CDC, AAP, and other experts recommend routine universal prenatal culture-based screening for GBS colonization (vaginal and rectal cultures) in all pregnant women at 35–37 weeks of gestation, unless GBS bacteriuria is known to be present during the current pregnancy or the woman had a previous infant with invasive GBS disease.359 These experts state that anti-infective prophylaxis to prevent perinatal GBS disease is indicated in pregnant women identified as GBS carriers during the routine prenatal GBS screening performed at 35–37 weeks during the current pregnancy, in women with GBS bacteriuria identified at any time during the current pregnancy, and in women who had a previous infant diagnosed with invasive GBS disease.359 Anti-infective prophylaxis to prevent perinatal GBS disease also is indicated in women with unknown GBS status at the time of onset of labor (i.e., culture not done, incomplete, or results unknown) if delivery is at less than 37 weeks of gestation, the duration of amniotic membrane rupture is 18 hours or longer, intrapartum temperature is 38°C or higher, or an intrapartum NAAT was positive for GBS.359

When intrapartum prophylaxis is indicated in the mother to prevent GBS disease in the neonate, it should be given at onset of labor or rupture of membranes.359 CDC, AAP, and other experts recommend penicillin G (5 million units IV initially followed by 2.5–3 million units IV every 4 hours until delivery) as the regimen of choice and ampicillin (2 g IV initially followed by 1 g IV every 4 hours until delivery) as the preferred alternative.359 If intrapartum GBS prophylaxis is indicated in a penicillin-allergic woman who is not at high risk for anaphylaxis (i.e., does not have a history of anaphylaxis, angioedema, respiratory distress, or urticaria after receiving a penicillin or cephalosporin), CDC, AAP, and others recommend IV cefazolin (2 g IV initially followed by 1 g IV every 8 hours until delivery).359 If intrapartum prophylaxis is indicated in a penicillin-allergic woman who is at high risk for anaphylaxis (e.g., history of anaphylaxis, angioedema, respiratory distress, or urticaria after receiving a penicillin or cephalosporin), IV clindamycin (900 mg IV every 8 hours until delivery) is recommended if the GBS isolate is susceptible to the drug;359 alternatively, IV vancomycin (1 g IV every 12 hours until delivery) can be used if the isolate is resistant to clindamycin.359

For additional information regarding prevention of perinatal group B streptococcal disease, the current CDC guidelines available at [Web] should be consulted.359

Cephalosporins General Statement Dosage and Administration

Administration

Cephalosporins, in appropriate forms, may be administered orally, IV, by deep IM injection, or intraperitoneally. In general, orally administered cephalosporins should not be relied on for the treatment of the initial phase of severe infections or in patients with nausea and vomiting. Cephalosporins should be given IV in patients with meningitis, septicemia, endocarditis, or other severe or life-threatening infections. Cephalosporins have been administered by regional perfusion, subconjunctivally, intraventricularly, or intrathecally, but the risk of CNS toxicity must be considered with the latter route. (See Cautions: Other Adverse Effects.)

Dosage

The duration of cephalosporin therapy depends on the type of infection. Generally, therapy should be continued for a minimum of 48–72 hours after the patient becomes asymptomatic or evidence of eradication of the infection has been obtained. In infections caused by β-hemolytic streptococci, therapy should be continued for at least 10 days. At least 4–6 weeks of therapy may be required in serious infections such as septicemia, endocarditis, or osteomyelitis.

Dosage in Renal Impairment

In patients with impaired renal function, decreases in doses and/or frequency of administration of cephalosporins may be required and should be based on the degree of renal impairment, severity of the infection, susceptibility of the causative organism, and serum concentrations of the cephalosporins.

Cautions for Cephalosporins General Statement

Cephalosporins generally are well tolerated and with only a few exceptions, adverse effects reported with the various cephalosporin derivatives are similar.351 The most frequent adverse effects reported with oral cephalosporins include GI effects (diarrhea, nausea, vomiting), headache, and rash and the most frequent adverse effects reported with parenteral cephalosporins include local reactions at the injection site, adverse GI effects, and adverse hematologic effects.351 Although not reported with other cephalosporins, some previously available cephalosporins (e.g., cefamandole, cefoperazone) and structurally related cephamycins (e.g., cefotetan) that contain an N-methylthiotetrazole (NMTT) side chain have been associated with an increased risk of hypoprothrombinemia and disulfiram-like reactions.309 312 313 390 In addition, while the clinical importance is unclear, derivatives that contain an NMTT side chain have caused adverse testicular effects in animal studies.309 312 313 353

Dermatologic and Sensitivity Reactions

Hypersensitivity reactions have been reported in approximately 5% or less of patients receiving a cephalosporin.351 These reactions include urticaria, pruritus, rash (maculopapular, erythematous, or morbilliform), fever and chills, eosinophilia, joint pain or inflammation, edema, facial edema, erythema, genital and anal pruritus, angioedema, shock, hypotension, vasodilatation, Stevens-Johnson syndrome, erythema multiforme, toxic epidermal necrolysis, and exfoliative dermatitis. Anaphylaxis, including a few fatalities, has occurred rarely with cephalosporins.

Serum sickness-like reactions consisting of erythema multiforme or maculopapular pruritic rash or urticaria accompanied by arthritis, arthralgia, and fever have been reported rarely in patients receiving cefaclor.307 351 427 428 429 430 431 These reactions usually have occurred in pediatric patients younger than 6 years of age, most often with second or subsequent courses of the drug.307 426 430 431 While serum sickness-like reactions have been reported rarely in patients receiving other cephalosporins (i.e., cefprozil, cephalexin) or other β-lactam antibiotics (i.e., amoxicillin, loracarbef),341 427 428 431 432 these reactions been reported more frequently with cefaclor than with any other anti-infective agent.427 428 429

Hematologic Effects

Positive direct and indirect antiglobulin (Coombs’) test results have been reported in 3% or more of patients receiving a cephalosporin. (See Laboratory Test Interferences: Immunohematology Tests.) The mechanism of this reaction is usually nonimmunologic in nature; a cephalosporin-globulin complex coats the erythrocytes and reacts nonspecifically with Coombs’ serum. Nonimmunologic positive Coombs’ test results are most likely to occur in patients who have received large doses of a cephalosporin or who have impaired renal function or hypoalbuminemia.

Other adverse hematologic effects of cephalosporins include rare, mild and transient neutropenia, thrombocythemia or thrombocytopenia, leukocytosis, granulocytosis, monocytosis, lymphocytopenia, basophilia, and reversible leukopenia. Transient lymphocytosis has been reported occasionally in infants and children receiving cefaclor. Rarely, decreased hemoglobin and/or hematocrit, anemia, and agranulocytosis have been reported with some cephalosporins. Aplastic anemia, pancytopenia, hemolytic anemia, and epistaxis or hemorrhage also have been reported with cephalosporin therapy. Immune-mediated hemolytic anemia with extravascular hemolysis, including some fatalities, have been reported in patients receiving cefotaxime,627 ceftriaxone,626 or cefotetan.628

Prolonged prothrombin time (PT), prolonged activated partial thromboplastin time (APTT), and/or hypoprothrombinemia (with or without bleeding) have been reported rarely with cefaclor, cefixime, cefoperazone (no longer available in the US), cefotaxime, cefotetan, cefoxitin, ceftizoxime (no longer commercially available in the US), ceftriaxone, and cefuroxime.316 317 318 319 320 321 323 324 343 677 Although the true incidence of hypoprothrombinemia and bleeding complications during therapy with these drugs has not been established, these effects have been reported most frequently with drugs that contain an NMTT side chain (e.g., cefamandole, cefoperazone [drugs no longer commercially available in the US]) and have usually occurred in geriatric, debilitated, or other patients with vitamin K deficiency or in patients with severe renal failure or following radical GI surgery.316 317 318 319 320 321 323 324 The mechanism(s) of the hypoprothrombinemic effect of these drugs has not been clearly established.316 317 318 319 320 321 323 324 While hypoprothrombinemia with some of the drugs may result in part from a reduction in vitamin K-producing bacteria in the GI tract,316 317 318 319 320 321 323 324 there is evidence that a direct effect on hepatic synthesis of prothrombin or prothrombin precursors is involved.316 317 318 319 320 321 323 324 The NMTT side chain apparently interferes with vitamin K metabolism and regeneration and inhibits γ-carboxylation of glutamic acid, the vitamin K-dependent step in the hepatic synthesis of prothrombin.316 320 321 324 Hypoprothrombinemia has usually been reversed by administration of vitamin K. Vitamin K should be administered when indicated to treat hypoprothrombinemia associated with use of other cephalosporins.

Renal and Genitourinary Effects

Renal effects that have occurred occasionally with administration of a cephalosporin include transient increases in BUN and serum creatinine concentrations, renal dysfunction, and toxic nephropathy. Nephrotoxicity has been reported rarely with cephalexin and cefazolin and acute renal failure has been reported with cefazolin310 and cefixime.677 Reversible interstitial nephritis has occurred rarely during cefaclor therapy, and purpuric nephritis has been reported with oral cefpodoxime proxetil therapy in postmarketing experience outside the US. Renal toxicity is most likely to occur in patients older than 50 years of age, patients with prior renal impairment, or patients who are receiving other nephrotoxic drugs. (See Drug Interactions: Nephrotoxic Drugs.) All cephalosporins should be administered with caution and in reduced dosage in the presence of markedly impaired renal function. In patients with suspected renal impairment, careful clinical observation and renal function tests should be performed prior to and during cephalosporin therapy.

Genitourinary effects reported with cephalosporin therapy include vaginitis, vaginal candidiasis, genital pruritus, and menstrual irregularities.

Hepatic Effects

Transient increases in serum AST (SGOT), ALT (SGPT), γ-glutamyl transferase (γ-glutamyl transpeptidase, GGT, GGTP), and alkaline phosphatase concentrations have occurred occasionally with cephalosporin therapy. Increased serum concentrations of bilirubin and/or LDH have been reported with many cephalosporins; decreased serum albumin and/or total protein also have been reported. Hepatic dysfunction, including cholestasis, also has been reported with cephalosporin therapy. Hepatitis and/or jaundice have been reported with cefazolin,310 cefixime,677 and ceftazidime.340 These hepatic effects are generally mild and disappear when cephalosporin therapy is discontinued.

Acute liver injury has been reported with oral cefpodoxime proxetil therapy in postmarketing experience outside the US.

GI Effects

The most frequent adverse reactions to orally administered cephalosporins are nausea, vomiting, and diarrhea. These effects are usually mild and transient, but rarely may be severe enough to require discontinuance of the drug. Other adverse GI effects that have occurred with some of the oral cephalosporins include abdominal pain, tenesmus, epigastric pain/dyspepsia, decreased appetite/anorexia, glossitis, flatulence, candidiasis (e.g., oral thrush), taste alteration, decreased salivation, and heartburn. Adverse GI effects also can occur with IM or IV cephalosporins.

Treatment with anti-infectives alters the normal flora of the colon leading to overgrowth of Clostridium difficile.302 307 308 310 311 314 332 339 340 343 347 348 349 677 703 C. difficile infection (CDI) and C. difficile-associated diarrhea and colitis (CDAD; also known as antibiotic-associated diarrhea and colitis or pseudomembranous colitis) have been reported with nearly all systemic anti-infectives, including cephalosporins, and may range in severity from mild diarrhea to fatal colitis.301 302 303 304 305 307 308 310 311 314 332 339 340 343 347 348 349 677 703 C. difficile produces toxins A and B which contribute to the development of CDAD;302 303 308 310 311 314 332 339 340 343 347 348 349 677 703 hypertoxin-producing strains of C. difficile are associated with increased morbidity and mortality since they may be refractory to anti-infectives and colectomy may be required.308 310 311 314 332 339 340 343 347 348 349 677 703

CDAD should be considered in the differential diagnosis in patients who develop diarrhea during or after anti-infective therapy.301 302 303 304 305 307 308 310 311 314 332 339 340 343 347 348 349 677 703 Careful medical history is necessary since CDAD has been reported to occur as late as 2 months or longer after anti-infective therapy is discontinued.308 310 311 314 332 339 340 343 347 348 349 677 703 If CDAD is suspected or confirmed, anti-infective therapy not directed against C. difficile should be discontinued whenever possible.301 302 304 305 308 310 311 314 332 339 340 343 347 348 349 677 703 Patients should be managed with appropriate supportive therapy (e.g., fluid and electrolyte management, protein supplementation), anti-infective therapy directed against C. difficile (e.g., metronidazole, vancomycin), and surgical evaluation as clinically indicated.301 302 303 304 305 308 310 311 314 332 339 340 343 347 348 349 677 703 Antiperistaltic agents (e.g., opiates, diphenoxylate with atropine) should be avoided since they may obscure symptoms and precipitate toxic megacolon.302 305

Local Effects

Local reactions are quite common following IM or IV administration of some parenteral cephalosporins; phlebitis and thrombophlebitis occasionally occur with IV administration of the drugs. Although reports are conflicting and results of many studies inconclusive, there appears to be little difference in the overall incidence of mild phlebitis and thrombophlebitis among the currently available IV cephalosporins.

Nervous System Effects

Nervous system effects that have occurred following oral, IM, or IV administration of cephalosporins include dizziness, headache, malaise, fatigue, nightmares, and vertigo. Hyperactivity, nervousness or anxiety, agitation, hallucinations, insomnia, somnolence, weakness, hot flushes, alteration in color perception, confusion, and hypertonia also have been reported during therapy with some cephalosporins, although a causal relationship has not necessarily been established. Mild to moderate hearing loss has been reported in a few pediatric patients receiving cefuroxime sodium for the treatment of meningitis.

In patients with renal insufficiency, elevated concentrations of ceftazidime reportedly may lead to seizures, encephalopathy, asterixis, and neuromuscular excitability. Seizures also have been reported with cefixime677 and cefuroxime.347 Rarely, toxic paranoid reactions have occurred in patients with renal impairment receiving oral cephalexin. Life-threatening or fatal encephalopathy (disturbance of consciousness including confusion, hallucinations, stupor, and coma), myoclonus, seizures, and nonconvulsive status epilepticus have been reported in patients receiving cefepime.314 Most cases of cefepime-associated neurotoxicity have occurred in patients with renal impairment who received a cefepime dosage that exceeded the recommended dosage for such patients; however, some cases occurred in patients who received a dosage appropriately adjusted for renal impairment.314

Intrathecal administration of cephalosporins, particularly in large doses, has resulted in CNS toxicity evidenced by hallucinations, nystagmus, and seizures. (See Cautions: Precautions and Contraindications.)

Other Adverse Effects

Other adverse effects reported with cephalosporin therapy include chest pain, pleural effusion, pulmonary infiltrate, dyspnea or respiratory distress, cough, and rhinitis. Increased or decreased serum glucose concentration also has been reported. Fungal skin infection, exacerbation of acne, and ocular itching have been reported rarely in patients receiving oral cefpodoxime proxetil therapy.

An anamnestic photosensitivity (photo recall)-like dermatitis, characterized by a pruritic, erythematous, maculopapular eruption distributed in the area of a recent sunburn on the upper abdomen and chest, has been reported in at least one patient receiving concomitant IV therapy with cefazolin and gentamicin sulfate. Whether this phenomenon was caused by one or both drugs has not been determined; however, the reaction resolved within 48 hours following discontinuance of both drugs.

Precautions and Contraindications

Prior to initiation of cephalosporin therapy, careful inquiry should be made concerning previous hypersensitivity reactions to cephalosporins, penicillins, and other drugs. Cephalosporins are contraindicated in patients with a history of allergic reactions to cephalosporin antibiotics. There is clinical and laboratory evidence of partial cross-sensitivity among bicyclic β-lactam antibiotics including penicillins, cephalosporins, cephamycins, and carbapenems.310 316 325 352 There appears to be little cross-sensitivity between bicyclic β-lactam antibiotics and monobactams (e.g., aztreonam).316 325 326 327 328 Although the true incidence of cross-sensitivity among the β-lactam antibiotics has not been definitely established,316 325 352 it has been clearly documented and may occur in up to 10–15% of patients with a history of penicillin hypersensitivity.292 310 339 350 Some patients have had severe reactions, including anaphylaxis, to both penicillins and cephalosporins.350 Cephalosporins should be used with caution in individuals hypersensitive to penicillins.310 338 339 350 Some clinicians suggest that cephalosporins should be avoided in patients who have had an immediate-type (anaphylactic) hypersensitivity reaction to penicillins and should be administered with caution to patients who have had a delayed-type (e.g., rash, fever, eosinophilia) reaction to penicillins or other drugs.292 351 If an allergic reaction occurs during cephalosporin therapy, the drug should be discontinued and the patient treated with appropriate therapy (e.g., epinephrine, corticosteroids, and maintenance of an adequate airway and oxygen) as indicated.

To reduce development of drug-resistant bacteria and maintain effectiveness of cephalosporins and other antibacterials, the drugs should be used only for the treatment or prevention of infections proven or strongly suspected to be caused by susceptible bacteria.308 310 340 When selecting or modifying anti-infective therapy, results of culture and in vitro susceptibility testing should be used.308 310 340 In the absence of such data, local epidemiology and susceptibility patterns should be considered when selecting anti-infectives for empiric therapy.308 310 340

Patients should be advised that antibacterials (including cephalosporins) should only be used to treat bacterial infections and not used to treat viral infections (e.g., the common cold).308 310 340 Patients also should be advised about the importance of completing the full course of therapy, even if feeling better after a few days, and that skipping doses or not completing therapy may decrease effectiveness and increase the likelihood that bacteria will develop resistance and will not be treatable with cephalosporins or other antibacterials in the future.308 310 340

Prolonged use of a cephalosporin may result in the overgrowth of nonsusceptible organisms, especially Enterobacter, Pseudomonas, enterococci, or Candida. If superinfection occurs, appropriate therapy should be instituted.

Cephalosporins should be used with caution in patients with a history of GI disease, particularly colitis.310 339 340 343 Because C. difficile-associated diarrhea and colitis has been reported with the use of cephalosporins, it should be considered in the differential diagnosis of patients who develop diarrhea during or following therapy with the drugs.302 310 339 340 343 (See Cautions: GI Effects.) Patients should be advised that diarrhea is a common problem caused by anti-infectives and usually ends when the drug is discontinued; however, they should contact a clinician if watery and bloody stools (with or without stomach cramps and fever) occur during or as late as 2 months or longer after the last dose.310 339 343

Seizures have been reported with several cephalosporins (e.g., ceftazidime, cefuroxime, cefepime), particularly in patients with renal impairment in whom dosage of the drug was not reduced.311 314 332 338 347 348 (See Cautions: Nervous System Effects.) If seizures occur during cephalosporin therapy, the drug should be discontinued and anticonvulsant therapy initiated as clinically indicated.311 314 332 338 347 348 If neurotoxicity associated with cefepime therapy occurs, consideration should be given to discontinuing the drug or making appropriate dosage adjustments based on the patient's renal function.314

Because some cephalosporins have been associated with a decrease in prothrombin activity, some manufacturers state that prothrombin time (PT) should be monitored when these drugs are used in patients with renal or hepatic impairment, in patients with poor nutritional status, in patients receiving a protracted course of anti-infective therapy, or in those previously stabilized on anticoagulant therapy.310 343 347 348 Vitamin K should be administered if indicated.310 343 347 348

Geriatric Precautions

No overall differences in safety and efficacy have been reported with use of cephalosporins in those 60 years of age and older compared with younger adults,307 308 310 314 332 339 340 Although clinical experience has revealed no evidence of age-related differences, the possibility of increased sensitivity in some geriatric individuals cannot be ruled out.307 308 310 332 339 340

Cephalosporins are substantially eliminated in urine and the risk of toxicity may be increased in patients with impaired renal function.307 308 310 314 332 339 340 Because geriatric patients are more likely to have decreased renal function, use caution when selecting dosage for such patients and consider monitoring renal function.307 308 310 314 332 339 340

Pregnancy, Fertility, and Lactation

Pregnancy

Although there have been no reports of adverse effects to the fetus to date, safe use of cephalosporins during pregnancy has not been definitely established. The drugs should be used during pregnancy only when clearly needed.

Fertility

Adverse testicular effects (e.g., reduced testicular weight, seminiferous tubule degeneration, delayed maturity of germinal epithelium, reduced germinal cell population, vacuolation of Sertoli cell cytoplasm) have been reported in prepubertal rats receiving certain previously available cephalosporins (e.g., cefamandole, cefoperazone) and structurally related cephamycins (e.g., cefotetan) that contain an NMTT side chain;309 312 313 314 315 impaired fertility also has been reported, particularly with high dosages.309 313 315 The relevance of these findings to humans is not known.309 312 313 314 315

Lactation

Because cephalosporins are distributed into milk, the drugs should be used with caution in nursing women.

Drug Interactions

Nephrotoxic Drugs

Concomitant use of nephrotoxic agents such as aminoglycosides, colistin, polymyxin B, or vancomycin may increase the risk of nephrotoxicity with some cephalosporins and probably should be avoided, if possible.

Alcohol

Disulfiram-like reactions have occurred when alcohol was ingested within 48–72 hours after administration of some previously available cephalosporins (e.g., cefamandole, cefoperazone) and structurally related cephamycins (e.g., cefotetan) that contain an N-methylthiotetrazole (NMTT) side chain.309 312 313 The reactions appear to result from accumulation of acetaldehyde and do not occur if alcohol is ingested prior to the first dose of the antibiotic.309 312 313

Estrogens or Progestins

Some cephalosporins (e.g., ceftazidime) may affect gut flora, leading to lower estrogen reabsorption and reduced efficacy of oral contraceptives containing estrogen and progesterone.340

Probenecid

Concomitant administration of oral probenecid competitively inhibits tubular secretion resulting in higher and more prolonged serum concentrations of most cephalosporins.

Other Anti-infective Agents

In vitro studies indicate that the antibacterial activity of cephalosporins may be additive or synergistic with aminoglycosides or penicillins against some organisms. Although some in vitro studies showed additive or synergistic antibacterial activity between chloramphenicol and a cephalosporin, there is more recent in vitro evidence of antagonism between cephalosporins (e.g., cefotaxime, ceftazidime, ceftriaxone) and chloramphenicol against a variety of gram-negative and -positive bacteria, particularly when chloramphenicol was added to the medium before the β-lactam. In addition, at least one case of in vivo antagonism has been reported in an infant with Salmonella enteritidis meningitis. Therefore, it is recommended that combined therapy with chloramphenicol and a cephalosporin be avoided, particularly when bactericidal activity is considered important.

Laboratory Test Interferences

Immunohematology Tests

Positive direct and indirect antiglobulin (Coombs’) test results have been reported in 3% or more of patients receiving a cephalosporin. (See Cautions: Hematologic Effects.) This reaction may interfere with hematologic studies or transfusion cross-matching procedures. In addition, positive Coombs’ test results may occur in neonates whose mother received a cephalosporin prior to delivery.

Tests for Urinary Glucose

Cephalosporins can cause false-positive results in urine glucose determinations using cupric sulfate solution (Benedict’s reagent, Clinitest); glucose oxidase tests (Clinistix) are unaffected by the drugs.

Tests for Creatinine

Some cephalosporins, in high concentrations, may cause falsely elevated serum or urine creatinine values when the Jaffé reaction is used. In one in vitro study, high concentrations of ceftriaxone (50 mcg/mL or greater) caused falsely elevated serum creatinine values when a manual method was used; however, other studies indicate that the drug does not interfere with automated methods for determining serum or urinary creatinine concentrations.

Mechanism of Action

Cephalosporins are usually bactericidal in action. The antibacterial activity of the cephalosporins, like penicillins and cephamycins, results from inhibition of mucopeptide synthesis in the bacterial cell wall. Although the exact mechanisms of action of cephalosporins have not been fully elucidated, β-lactam antibiotics bind to several enzymes in the bacterial cytoplasmic membrane (e.g., carboxypeptidases, endopeptidases, transpeptidases) that are involved in cell-wall synthesis and cell division. It has been hypothesized that β-lactam antibiotics act as substrate analogs of acyl-d-alanyl-d-alanine, the usual substrate for these enzymes. This interferes with cell-wall synthesis and results in the formation of defective cell walls and osmotically unstable spheroplasts. Cell death following exposure to β-lactam antibiotics usually results from lysis, which appears to be mediated by bacterial autolysins such as peptidoglycan hydrolases.

The target enzymes of β-lactam antibiotics have been classified as penicillin-binding proteins (PBPs) and appear to vary substantially among bacterial species. The affinities of various β-lactam antibiotics for different PBPs appear to explain the differences in morphology that occur in susceptible organisms following exposure to different β-lactam antibiotics and may also explain differences in the spectrum of activity of β-lactam antibiotics that are not caused by the presence or absence of β-lactamases.

Spectrum

In general, cephalosporins are active in vitro against many gram-positive aerobic bacteria, some gram-negative aerobic bacteria, and some anaerobic bacteria; however, there are substantial differences among the cephalosporins in spectra of activity as well as levels of activity against susceptible bacteria. Cephalosporins are inactive against fungi and viruses.

Classification of Cephalosporins and Closely Related β-Lactam Antibiotics Based on Spectra of Activity

Currently available cephalosporins are generally divided into 5 groups based on their spectra of activity: first, second, third, fourth, and fifth generation cephalosporins. Closely related β-lactam antibiotics (e.g., cephamycins) also may be classified in these groups because of their similar spectra of activity.

First Generation Cephalosporins

First generation cephalosporins usually are active in vitro against gram-positive cocci including penicillinase-producing and nonpenicillinase-producing Staphylococcus aureus and S. epidermidis; Streptococcus pyogenes (group A β-hemolytic streptococci); S. agalactiae (group B streptococci); and S. pneumoniae. First generation cephalosporins have limited activity against gram-negative bacteria, although some strains of Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, and Shigella may be inhibited in vitro by the drugs. First generation cephalosporins are inactive against enterococci, including Enterococcus faecalis (formerly S. faecalis), methicillin-resistant S. aureus (MRSA; also known as oxacillin-resistant S. aureus, ORSA), Bacteroides fragilis, Citrobacter, Enterobacter, Listeria monocytogenes, Proteus other than P. mirabilis, Providencia, Pseudomonas, and Serratia.

Susceptible strains of S. aureus, S. pneumoniae, S. pyogenes, or S. agalactiae usually are inhibited in vitro by cefazolin concentrations of 0.1–1 mcg/mL or cephalexin concentrations of 0.1–12 mcg/mL. S. epidermidis may be inhibited in vitro by cefazolin concentrations of 0.1–12.5 mcg/mL, although some strains require concentrations greater than 32 mcg/mL for in vitro inhibition. Susceptible strains of E. coli, K. pneumoniae, or P. mirabilis generally are inhibited in vitro by cefazolin concentrations of 0.8–12.5 mcg/mL.

Second Generation Cephalosporins

Second generation cephalosporins usually are active in vitro against bacteria susceptible to first generation cephalosporins. In addition, the second generation drugs are active in vitro against most strains of Haemophilus influenzae (including ampicillin-resistant strains). Although the specific spectra of activity differ, second generation cephalosporins generally are more active in vitro against gram-negative bacteria than first generation cephalosporins. However, cefaclor is less active than other second generation cephalosporins against gram-negative bacteria. The second generation drugs may be active in vitro against some strains of Acinetobacter, Citrobacter, Enterobacter, E. coli, Klebsiella, Neisseria, Proteus, Providencia, and Serratia that are resistant to the first generation drugs. Cefoxitin also has some activity in vitro against B. fragilis. Second generation cephalosporins are inactive against enterococci (e.g., E. faecalis), MRSA, and Pseudomonas.

Third Generation Cephalosporins

Third generation cephalosporins usually are less active in vitro against susceptible staphylococci than first generation cephalosporins; however, the third generation drugs have an expanded spectrum of activity against gram-negative bacteria compared with the first and second generation drugs. Third generation cephalosporins generally are active in vitro against gram-negative bacteria susceptible to the first and second generation drugs, and most also are active in vitro against some strains of Citrobacter, Enterobacter, E. coli, Klebsiella, Neisseria, Proteus, Morganella, Providencia, and Serratia that may be resistant to first and second generation cephalosporins. Cefdinir, cefixime, and cefpodoxime are inactive against most strains of Enterobacter and Pseudomonas and have limited in vitro activity against anaerobic bacteria; cefixime also is inactive against most staphylococci. Cefditoren has increased activity, similar to first generation cephalosporins, against gram-positive bacteria, unlike other third generation cephalosporins.664 Third generation cephalosporins are inactive against MRSA and generally are inactive against enterococci (e.g., E. faecalis) and L. monocytogenes. Some parenteral third generation cephalosporins (ceftazidime) have activity in vitro against Ps. aeruginosa.715

Fourth Generation Cephalosporins

Fourth generation cephalosporins, like third generation cephalosporins, have an expanded spectrum of activity against gram-negative bacteria compared with the first and second generation drugs.383 384 385 However, fourth generation cephalosporins are active in vitro against some gram-negative bacteria, including Pseudomonas aeruginosa and certain Enterobacteriaceae, that generally are resistant to third generation cephalosporins.383 384 385 In addition, fourth generation cephalosporins may be more active against gram-positive bacteria than some third generation drugs (e.g., ceftazidime).383 384 385 387 Cefepime has a spectrum of activity against aerobic gram-positive and gram-negative bacteria that is similar to that of cefotaxime and ceftriaxone and has activity against Ps. aeruginosa that appears to approach that of ceftazidime.384 385 387 However, cefepime is more active than third generation cephalosporins against Enterobacteriaceae that produce inducible β-lactamases.383 384 386 The extended spectrum of activity of cefepime is related to the fact that the drug penetrates the outer membrane of gram-negative bacteria more rapidly than most other cephalosporins and the fact that the drug is more resistant to inactivation by chromosomally and plasmid-mediated β-lactamases than most other cephalosporins.383 384 385 386 387 388 In addition, inducible β-lactamases have a low affinity for cefepime and the drug is hydrolyzed by these enzymes at a slower rate than third generation cephalosporins such as ceftazidime.383 387 Cefepime is inactive against MRSA, enterococci, and L. monocytogenes.383 384 386 387

Fifth Generation Cephalosporins

Like third and fourth generation cephalosporins, fifth generation cephalosporins have an expanded spectrum of activity that includes both gram-positive and gram-negative bacteria.704 705 711 712 713 714 715 Fifth generation cephalosporins also usually have activity against methicillin-resistant Staphylococcus aureus (MRSA; also known as oxacillin-resistant S. aureus, ORSA).704 705 711 712 713 715

In Vitro Susceptibility Testing

When in vitro susceptibility testing is performed according to the standards of the Clinical and Laboratory Standards Institute (CLSI; formerly National Committee for Clinical Laboratory Standards [NCCLS]), clinical isolates identified as susceptible to a cephalosporin are inhibited by drug concentrations usually achievable when the recommended dosage is used for the site of infection.676 Clinical isolates classified as intermediate have minimum inhibitory concentrations (MICs) that approach usually attainable blood and tissue concentrations and response rates may be lower than for strains identified as susceptible.676 Therefore, the intermediate category implies clinical applicability in body sites where the drug is physiologically concentrated or when a higher than usual dosage can be used.676 This intermediate category also includes a buffer zone which should prevent small, uncontrolled technical factors from causing major discrepancies in interpretation, especially for drugs with narrow pharmacotoxicity margins.676 If results of in vitro susceptibility testing indicate that a clinical isolate is resistant to a cephalosporin, the strain is not inhibited by drug concentrations generally achievable with usual dosage schedules and/or MICs fall in the range where specific microbial resistance mechanisms are likely and clinical efficacy of the drug against the isolate has not been reliably demonstrated in clinical studies.676

Because of differences in spectra of activity, the first generation cephalosporins (cefazolin), second generation cephalosporins (cefaclor, cefprozil, cefuroxime), third generation cephalosporins (cefdinir, cefditoren, cefixime, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone), fourth generation cephalosporins (cefepime), and fifth generation cephalosporins (ceftaroline) should be tested individually to determine in vitro susceptibility.676

CLSI states that in vitro susceptibility test results for cephalothin (no longer commercially available in the US) should only be used to predict susceptibility of Enterobacteriaceae to certain oral cephalosporins (i.e., cefadroxil, cephalexin, cefpodoxime).676 Although older data suggest that cephalothin results could predict susceptibility to some other cephalosporins, recent data are not available to confirm this.676

Disk Susceptibility Tests

When the disk-diffusion procedure is used to test in vitro susceptibility, individual disks containing cefaclor (30 mcg), cefazolin (30 mcg), cefdinir (5 mcg), cefditoren (5 mcg), cefepime (30 mcg), cefixime (5 mcg), cefotaxime (30 mcg), cefotetan (30 mcg), cefoxitin (30 mcg), cefpodoxime (10 mcg), cefprozil (30 mcg), ceftaroline (30 mcg), ceftazidime (30 mcg), ceftibuten (30 mcg), ceftriaxone (30 mcg), or cefuroxime (30 mcg) should be used.676 There is no class susceptibility disk that can be used to test susceptibility to all cephalosporins.676

The disk-diffusion procedure should not be used to test in vitro susceptibility of Streptococcus pneumoniae to cephalosporins; in vitro susceptibility should be evaluated using broth or agar dilutions tests.676

For detailed information on interpretation of disk diffusion zone diameters for disk susceptibility tests performed according to CLSI standardized procedures, see specialized references.676

Dilution Susceptibility Tests

When broth or agar dilution susceptibility tests are used to test in vitro susceptibility to cephalosporins, each drug must be tested individually.676

For detailed information on interpretation of MICs (mcg/mL) for diffusion susceptibility tests performed according to CLSI standardized procedures, see specialized references.676

Resistance

Bacterial resistance to cephalosporins may be natural or acquired and may result from one or a combination of factors. Some bacteria are not affected by concentrations of a cephalosporin that are lethal to other bacteria because their cell surfaces are not permeable to the drug or because their metabolic pathways are not inhibited by the drug. A major mechanism of bacterial resistance to cephalosporins is the production of β-lactamases which inactivate the drugs by hydrolyzing the β-lactam ring. However, absence or presence of a β-lactamase does not entirely dictate susceptibility or resistance to a cephalosporin. Bacterial resistance usually results both from the production of a β-lactamase and the presence of permeability barriers to the drug. The β-lactamases produced by different bacterial species differ in physical, chemical, and functional properties. Staphylococcal β-lactamases are usually inducible, extracellular penicillinases. A variety of β-lactamases are produced by gram-negative bacteria; some are chromosome-mediated inducible cephalosporinases and a few are determined by plasmid-mediated resistance factors.

Cephalosporins General Statement Pharmacokinetics

Absorption

Cefazolin sodium, cefepime hydrochloride, cefotaxime sodium, ceftaroline fosamil, ceftazidime, ceftriaxone sodium, and cefuroxime sodium are not appreciably absorbed from the GI tract and must be given parenterally.

Cefaclor, cefadroxil, cefprozil, ceftibuten dihydrate, cephalexin, and cephalexin hydrochloride are well absorbed from the GI tract. Following oral administration, bioavailability of cefdinir averages 16–25%. Approximately 30–50% of an oral dose of cefixime and 50% of an oral dose of cefpodoxime proxetil is absorbed from the GI tract. Cefditoren pivoxil,663 cefpodoxime proxetil,338 and cefuroxime axetil348 are prodrugs and are inactive until hydrolyzed in vivo to cefditoren, cefpodoxime, and cefuroxime, respectively, by nonspecific esterases in the intestinal lumen and blood. Ceftaroline fosamil also is a prodrug that is inactive until it is converted in vivo to ceftaroline by a plasma phosphatase following IV administration.703 704

Administration with food does not affect the rate of absorption or peak serum concentrations of cefadroxil. While the rate of absorption of cefixime or cefprozil may be decreased by the presence of food in the GI tract, the extent of absorption and peak plasma concentrations of the drug generally are not affected. Compared with administration in the fasting state, administration of cefdinir with a high-fat meal decreases the rate and extent of absorption of the drug. Administration of cefditoren pivoxil with a moderate- or high-fat meal increases the rate and extent of absorption, compared with administration in the fasting state.663 The effect of food on oral bioavailability of cefaclor, cefpodoxime proxetil, ceftibuten, and cefuroxime axetil varies depending on the formulation of the drugs administered.

Distribution and Elimination

Following absorption, most cephalosporins are widely distributed to tissues and fluids, including pleural fluid, synovial fluid, and bone. Following oral administration, cefdinir, cefixime, cefpodoxime, ceftibuten, cefprozil, and cefuroxime are distributed into middle ear fluid, tonsils, sinus tissue, and bronchial mucosa. Following oral administration, cefditoren has been shown to distribute into tonsils and skin blister fluid.663 Although the total quantity of some cephalosporins distributed into bile is low, therapeutic concentrations of some of the drugs (e.g., cefazolin, cefepime, cefixime) generally are obtained if biliary obstruction is not present. Only low concentrations of first or second generation cephalosporins diffuse into CSF following oral, IM, or IV administration even when meninges are inflamed; however, therapeutic concentrations of cefotaxime, ceftazidime, ceftriaxone, or cefuroxime generally are attained in CSF following IM or IV administration, especially if meninges are inflamed. Cefepime also is distributed into CSF following parenteral administration.

Cephalosporins readily cross the placenta, and fetal serum concentrations may be 10% or more of maternal serum concentrations. Cephalosporins are distributed in low concentrations into milk.

Serum cephalosporin concentrations may be higher and the serum half-lives prolonged in patients with impaired renal function.

Cefaclor, cefadroxil, cefazolin, cefdinir, cefditoren, cefixime, cefpodoxime, cefprozil, ceftazidime, ceftolozane, cefuroxime, and cephalexin are not appreciably metabolized. Cefuroxime axetil, cefditoren pivoxil, and cefpodoxime proxetil are rapidly hydrolyzed to their respective microbiologically active forms, cefuroxime, cefditoren, and cefpodoxime, by nonspecific esterases in the intestinal mucosa and/or blood following oral administration. The axetil moiety of cefuroxime is metabolized to acetaldehyde and acetic acid; hydrolysis of cefditoren pivoxil results in the formation of pivalate, which is absorbed and excreted as pivaloylcarnitine in urine. Cefepime is partially metabolized in vivo. Cefotaxime is partially metabolized (presumably in the liver, kidneys, and other tissues) to desacetyl metabolites which also have antibacterial activity, although less than that of the parent compounds. Ceftriaxone is metabolized to a small extent to microbiologically inactive metabolites in the intestines after biliary excretion.

Cephalosporins and their metabolites are rapidly excreted by the kidneys. Cefaclor, cefadroxil, cefazolin, cefditoren, cefixime, cefotaxime, cefpodoxime, cefprozil, cefuroxime, and cephalexin are excreted by both glomerular filtration and tubular secretion; cefepime, ceftazidime, ceftolozane, and ceftriaxone are excreted principally by glomerular filtration. The same system of anion transport is responsible for the tubular secretion of cephalosporins as for other β-lactam antibiotics and probenecid. Oral probenecid administered shortly before or with most cephalosporins usually slows the rate of excretion of the antibiotic and produces higher and more prolonged serum concentrations, especially with those cephalosporins excreted principally by tubular secretion. Most cephalosporins are removed by hemodialysis or peritoneal dialysis.

Chemistry and Stability

Chemistry

Cephalosporins are semisynthetic antibiotic derivatives of cephalosporin C, a substance produced by the fungus Cephalosporium acremonium. The drugs are β-lactam antibiotics structurally and pharmacologically related to penicillins and cephamycins (e.g., cefoxitin). All commercially available cephalosporins contain the 7-aminocephalosporanic acid (7-ACA) nucleus, which is composed of a β-lactam ring fused with a 6-membered dihydrothiazine ring instead of the 5-membered thiazolidine ring of penicillins. Cleavage at any point in the β-lactam ring system of cephalosporins results in complete loss of antibacterial activity.

Addition of various groups at R1 (position 7) and R2 (position 3) of the cephalosporin nucleus results in derivatives with differences in spectra of activity, stability against hydrolysis by β-lactamases, protein binding, GI absorption, or susceptibility to desacetylation. Many commercially available oral cephalosporins (e.g., cefdinir, cefditoren pivoxil, cefixime, cefpodoxime proxetil, ceftibuten) and parenteral cephalosporins (e.g., cefepime, cefotaxime, ceftazidime, ceftriaxone) contain an aminothiazolyl side chain at position 7 of the cephalosporin nucleus. The aminothiazolyl side chain enhances antibacterial activity, particularly against Enterobacteriaceae, and generally results in enhanced stability against β-lactamases. Unlike other commercially available cephalosporins, ceftaroline contains a 1,3-thiazole ring, linked to the 3-position of the cephem ring by a sulfur, which appears to contribute to activity against methicillin-resistant Staphylococcus aureus (MRSA; also known as oxacillin-resistant S. aureus or ORSA).704 Some previously available cephalosporins (e.g., cefamandole, cefoperazone) and structurally related cephamycins (e.g., cefotetan) contain an N-methylthiotetrazole (NMTT) side chain at position 6 of the cephalosporin nucleus.312 313 The NMTT side chain enhances antibacterial activity, helps to prevent metabolism of the drugs, and also may be associated with certain adverse effects (e.g., hypoprothrombinemia, disulfiram-like reactions).312 313 390

Stability

In solution, most cephalosporins are stable for only short periods of time unless frozen.

Cephalosporins are potentially physically and/or chemically incompatible with some drugs including aminoglycosides, but the compatibility depends on the specific drug and several other factors (e.g., concentration of the drugs, specific diluents used, resulting pH, temperature). Specialized references should be consulted for specific compatibility information.

Releated Monographs

For specific dosages and additional information on chemistry and stability, spectrum, resistance, pharmacokinetics, uses, cautions, drug interactions, and laboratory test interferences of the cephalosporins, see the individual monographs in 8:12.06. The American Society of Health-System Pharmacists, Inc. represents that the information provided in the accompanying monograph was formulated with a reasonable standard of care, and in conformity with professional standards in the field. Readers are advised that decisions regarding use of drugs are complex medical decisions requiring the independent, informed decision of an appropriate health care professional, and that the information contained in the monograph is provided for informational purposes only. The manufacturer’s labeling should be consulted for more detailed information. The American Society of Health-System Pharmacists, Inc. does not endorse or recommend the use of any drug. The information contained in the monograph is not a substitute for medical care.

AHFS DI Essentials™. © Copyright 2024, Selected Revisions September 14, 2023. American Society of Health-System Pharmacists, Inc., 4500 East-West Highway, Suite 900, Bethesda, Maryland 20814.

† Off-label: Use is not currently included in the labeling approved by the US Food and Drug Administration.

References

Only references cited for selected revisions after 1984 are available electronically.

173. Johnson RC, Bey RF, Wolgamot SJ. Comparison of the activities of ceftriaxone and penicillin G against experimentally induced syphilis in rabbits. Antimicrob Agents Chemother. 1982; 21:984-9. http://www.ncbi.nlm.nih.gov/pubmed/6287932?dopt=AbstractPlus

192. Chow AW, Benninger MS, Brook I et al. IDSA clinical practice guideline for acute bacterial rhinosinusitis in children and adults. Clin Infect Dis. 2012; 54:e72-e112. http://www.ncbi.nlm.nih.gov/pubmed/22438350?dopt=AbstractPlus

193. Wald ER, Applegate KE, Bordley C et al. Clinical Practice Guideline for the Diagnosis and Management of Acute Bacterial Sinusitis in Children Aged 1 to 18 Years. Pediatrics. 2013; :. http://www.ncbi.nlm.nih.gov/pubmed/23796742?dopt=AbstractPlus

216. Moorthy TT, Lee CT, Lim KB et al. Ceftriaxone for treatment of primary syphilis in men: a preliminary study. Sex Transm Dis. 1987; 14:116-8. http://www.ncbi.nlm.nih.gov/pubmed/3616852?dopt=AbstractPlus

228. Marra CM, Boutin P, McArthur JC et al. A pilot study evaluating ceftriaxone and penicillin G as treatment agents for neurosyphilis in human immunodeficiency virus-infected individuals. Clin Infect Dis. 2000; 30:540-4. http://www.ncbi.nlm.nih.gov/pubmed/10722441?dopt=AbstractPlus

260. Hook EW, Roddy RE, Handsfield HH. Ceftriaxone therapy for incubating and early syphilis. J Infect Dis. 1988; 158:881-4. http://www.ncbi.nlm.nih.gov/pubmed/3171231?dopt=AbstractPlus

292. American Academy of Pediatrics. Red Book: 2012 Report of the Committee on Infectious Diseases. 29th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2012.

301. Jaber MR, Olafsson S, Fung WL et al. Clinical review of the management of fulminant Clostridium difficile infection. Am J Gastroenterol. 2008; 103:3195-203; quiz 3204. http://www.ncbi.nlm.nih.gov/pubmed/18853982?dopt=AbstractPlus

302. Cohen SH, Gerding DN, Johnson S et al. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America (IDSA). Infect Control Hosp Epidemiol. 2010; 31:431-55. http://www.ncbi.nlm.nih.gov/pubmed/20307191?dopt=AbstractPlus

303. Fekety R for the American College of Gastroenterology Practice parameters Committee. Guidelines for the diagnosis and management of Clostridium difficile-associated diarrhea and colitis. Am J Gastroenterol. 1997; 92:739-50. http://www.ncbi.nlm.nih.gov/pubmed/9149180?dopt=AbstractPlus

304. American Society of Health-System Pharmacists Commission on Therapeutics ASHP therapeutic position statement on the preferential use of metronidazole for the treatment of Clostridium difficile-associated disease. Am J Health-Syst Pharm. 1998; 55:1407-11.

305. Bauer MP, Kuijper EJ, van Dissel JT et al. European Society of Clinical Microbiology and Infectious Diseases (ESCMID): treatment guidance document for Clostridium difficile infection (CDI). Clin Microbiol Infect. 2009; 15:1067-79. http://www.ncbi.nlm.nih.gov/pubmed/19929973?dopt=AbstractPlus

307. Alvogen Inc. Cefaclor capsules prescribing information. Parsippany, NJ; 2010 Aug.

308. West-ward Pharmaceutical Corp. Cefadroxil tablets, film coated and cefadroxil capsules prescribing information. Eatontown, NJ; 2011 Feb.

309. Lilly. Mandol (cefamandole nafate) for injection prescribing information (dated 1996 Oct 1). In: Physicians’ desk reference. 53rd ed. Montvale, NJ: Medical Economics Company Inc; 1999:1619-21.

310. Sandoz Inc. Cefazolin for injection, USP (500 mg and 1 g vials) prescribing information. Princeton, NJ; 2013 Feb.

311. Teva Pharmaceuticals USA. Cefdinir capsules prescribing information. Sellersville, PA; 2009 Dec.

312. Pfizer. Cefobid (sterile cefoperazone sodium) for intravenous or intramuscular use prescribing information (dated 1996 Oct). In: Physicians’ desk reference. 53rd ed. Montvale, NJ: Medical Economics Company Inc; 1999:2372-4.

313. Abraxis. Cefotetan for injection prescribing information. Schaumburg, IL; 2007 Jun.

314. Hospira. Maxipime (cefepime hydrochloride, USP) for injection for intravenous or intramuscular use prescribing information. Lake Forest, IL; 2013 Apr.

315. Manson JM, Zolna LE, Kang YJ et al. Effects of cefonicid and other cephalosporin antibiotics on male sexual development in rats. Antimicrob Agents Chemother. 1987; 31:991-7. http://www.ncbi.nlm.nih.gov/pubmed/3662478?dopt=AbstractPlus

316. Norrby SR. Adverse reactions and interactions with newer cephalosporin and cephamycin antibiotics. Medical Toxicol. 1986; 1:32-46.

317. Agnelli G, Del Favero A, Parise P et al. Cephalosporin-induced hypoprothrombinemia: is the N-methylthiotetrazole side chain the culprit? Antimicrob Agents Chemother. 1986; 29:1108-9.

318. Brown RB, Klar J, Lemeshow S et al. Enhanced bleeding with cefoxitin or moxalactam: statistical analysis within a defined population of 1493 patients. Arch Intern Med. 1986; 146:2159-64. http://www.ncbi.nlm.nih.gov/pubmed/3778044?dopt=AbstractPlus

319. Nichols RL, Wikler MA, McDevitt JT et al. Coagulopathy associated with extended-spectrum cephalosporins in patients with serious infections. Antimicrob Agents Chemother. 1987; 31:281-5. http://www.ncbi.nlm.nih.gov/pubmed/3471181?dopt=AbstractPlus

320. Barza M, Furie B, Brown AE et al. Defects in vitamin K-dependent carboxylation associated with moxalactam treatment. J Infect Dis. 1986; 153:1166-9. http://www.ncbi.nlm.nih.gov/pubmed/3701122?dopt=AbstractPlus

321. Andrassy K, Bechtold H, Ritz E. Hypoprothrombinemia caused by cephalosporins. J Antimicrob Chemother. 1985; 15:133-5. http://www.ncbi.nlm.nih.gov/pubmed/3980307?dopt=AbstractPlus

323. Barriere SL, Flaherty JF. Third-generation cephalosporins: a critical evaluation. Clin Pharm. 1984; 3:351-73. http://www.ncbi.nlm.nih.gov/pubmed/6432420?dopt=AbstractPlus

324. Sattler FR, Weitekamp MR, Ballard JO. Potential for bleeding with the new beta-lactam antibiotics. Ann Intern Med. 1986; 105:924-31. http://www.ncbi.nlm.nih.gov/pubmed/3535606?dopt=AbstractPlus

325. Saxon A, Beall GN, Rohr AS et al. Immediate hypersensitivity reactions to beta-lactam antibiotics. Ann Intern Med. 1987; 107:204-15. http://www.ncbi.nlm.nih.gov/pubmed/3300459?dopt=AbstractPlus

326. Adkinson NF, Swabb EA, Sugerman AA. Immunology of the monobactam aztreonam. Antimicrob Agents Chemother. 1984; 25:93-7. http://www.ncbi.nlm.nih.gov/pubmed/6538398?dopt=AbstractPlus

327. Adkinson N, Saxon A, Spence MR et al. Cross-allergenicity and immunogenicity of aztreonam. Rev Infect Dis. 1985; 7(Suppl 4):S613-21. http://www.ncbi.nlm.nih.gov/pubmed/4081473?dopt=AbstractPlus

328. Saxon A, Swabb EA, Adkinson NF. Investigation into the immunologic cross-reactivity of aztreonam with other beta-lactam antibiotics. Am J Med. 1985; 78(Suppl 2A):19-26. http://www.ncbi.nlm.nih.gov/pubmed/2578733?dopt=AbstractPlus

329. Wormser GP, Dattwyler RJ, Shapiro ED et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2006; 43:1089-134. http://www.ncbi.nlm.nih.gov/pubmed/17029130?dopt=AbstractPlus

330. Marchant CD, Shurin PA, Johnson CE et al. A randomized controlled trial of amoxicillin plus clavulanate compared with cefaclor for treatment of acute otitis media. J Pediatr. 1986; 109:891-6. http://www.ncbi.nlm.nih.gov/pubmed/3534203?dopt=AbstractPlus

331. Van Hare GF, Shurin PA, Marchant CD et al. Acute otitis media caused by Branhamella catarrhalis: biology and therapy. Rev Infect Dis. 1987; 9:16-27. http://www.ncbi.nlm.nih.gov/pubmed/3493519?dopt=AbstractPlus

332. Sanofi-Aventis. Claforan (cefotaxime sodium) injection and for injection prescribing information. Bridgewater, NJ; 2009 Jul.

333. Odio CM, Kusmiesz H, Shelton S et al. Comparative treatment trial of augmentin versus cefaclor for acute otitis media with effusion. Pediatrics. 1985; 75:819-26. http://www.ncbi.nlm.nih.gov/pubmed/4039433?dopt=AbstractPlus

334. Van Hare GF, Shurin PA. The increasing importance of Branhamella catarrhalis in respiratory infections. Pediatr Infect Dis J. 1987; 6:92-4. http://www.ncbi.nlm.nih.gov/pubmed/3547295?dopt=AbstractPlus

335. Doern GV. Branhamella catarrhalis—an emerging human pathogen. Diagn Microbiol Infect Dis. 1986; 4:191-201. http://www.ncbi.nlm.nih.gov/pubmed/3514103?dopt=AbstractPlus

336. Bluestone CD. Otitis media and sinusitis in children: role of Branhamella catarrhalis . Drugs. 1986; 31(Suppl 3):132-41. http://www.ncbi.nlm.nih.gov/pubmed/3732081?dopt=AbstractPlus

337. Kallings I. Sensitivity of Branhamella catarrhalis to oral antibiotics. Drugs. 1986; 31(Suppl 3):17-22. http://www.ncbi.nlm.nih.gov/pubmed/3488193?dopt=AbstractPlus

338. Sandoz. Cefpodoxime proxetil granule for suspension prescribing information. Princeton, NJ; 2008 Sep.

339. Lupin Pharmaceuticals Inc. Cefprozil tablets prescribing information. Baltimore, MD; 2007 Dec.

340. Covis Pharmaceuticals Inc. Fortaz (ceftazidime) powder for injection and injection prescribing information. Cary, NC; 2012 Apr.

341. Pernix. Cedax (ceftibuten dihydrate) capsules and oral suspension prescribing information. Gonzales, LA; 2010 Apr.

343. Genentech USA, Inc. Rocephin (ceftriaxone sodium) injection powder, for solution prescribing information. South San Francisco, CA; 2013 Apr.

344. Workowski KA, Bolan GA. Sexually Transmitted Diseases Treatment Guidelines, 2015. MMWR Recomm Rep. 2015; 64(RR-03):1-137. http://www.ncbi.nlm.nih.gov/pubmed/26042815?dopt=AbstractPlus

347. Covis Pharmaceutics Inc. Zinacef (cefuroxime sodium) powder for injection and injection prescribing information. Cary, NC; 2013 Jun.

348. GlaxoSmithKline. Ceftin (cefuroxime axetil) powder for oral suspension and film-coated tablets prescribing information. Research Triangle Park, NC; 2010 Jan.

349. Shionogi Inc. Keflex (cephalexin) capsules prescribing information. Florham Park, NJ; 2011 Aug.

350. Ranbaxy Pharmaceuticals Inc. Cephalexin capsules and cephalexin oral suspension prescribing information. Jacksonville, FL; 2007 Jan.

351. Thompson JW, Jacobs RF. Adverse effects of newer cephalosporins: an update. Drug Saf. 1993; 9:132-42. http://www.ncbi.nlm.nih.gov/pubmed/8397890?dopt=AbstractPlus

352. Kishiyama JL, Adelman DC. The cross-reactivity and immunology of β-lactam antibiotics. Drug Saf. 1994; 10:318-27. http://www.ncbi.nlm.nih.gov/pubmed/8018304?dopt=AbstractPlus

353. Manson JM, Zolna LE, Kang YJ et al. Effects of cefonicid and other cephalosporin antibiotics on male sexual development in rats. Antimicrob Agents Chemother. 1987; 31:991-7. http://www.ncbi.nlm.nih.gov/pubmed/3662478?dopt=AbstractPlus

354. Meijer WS, Schmitz PIM, Jeekel J. Meta-analysis of randomized, controlled clinical trials of antibiotic prophylaxis in biliary tract surgery. Br J Surg. 1990; 77:283-90. http://www.ncbi.nlm.nih.gov/pubmed/2138925?dopt=AbstractPlus

355. Classen DC, Evans RS, Pestotnik SL et al. The timing of prophylactic administration of antibiotics and the risk of surgical-wound infection. N Engl J Med. 1992; 326:281-6. http://www.ncbi.nlm.nih.gov/pubmed/1728731?dopt=AbstractPlus

356. Wenzel RP. Preoperative antibiotic prophylaxis. N Engl J Med. 1992; 326:337-9. http://www.ncbi.nlm.nih.gov/pubmed/1728738?dopt=AbstractPlus

357. Gorbach SL. The role of cephalosporins in surgical prophylaxis. J Antimicrob Chemother. 1989; 23(Suppl D):61-70. http://www.ncbi.nlm.nih.gov/pubmed/2656627?dopt=AbstractPlus

358. Lewis RT, Goodall RG, Marien B et al. Biliary bacteria, antibiotic use, and wound infection in surgery of the gallbladder and common bile duct. Arch Surg. 1987; 122:44-7. http://www.ncbi.nlm.nih.gov/pubmed/3800650?dopt=AbstractPlus

359. Verani JR, McGee L, Schrag SJ et al. Prevention of perinatal group B streptococcal disease--revised guidelines from CDC, 2010. MMWR Recomm Rep. 2010; 59(RR-10):1-36. http://www.ncbi.nlm.nih.gov/pubmed/21088663?dopt=AbstractPlus

360. . Antimicrobial prophylaxis for surgery. Treat Guidel Med Lett. 2012; 10:73-8; quiz 79-80. http://www.ncbi.nlm.nih.gov/pubmed/22996382?dopt=AbstractPlus

361. Fromtling RA, Gadebusch HH. Ethanol-cephalosporin antibiotic interactions: an animal model for the detection of disulfiram (Antabuse)-like effects. Methods Find Exp Clin Pharmacol. 1983; 5:595-600. http://www.ncbi.nlm.nih.gov/pubmed/6668968?dopt=AbstractPlus

362. Lilly, Indianapolis, IN: Personal communication.

363. Anon. Prevalence of penicillin-resistant Streptococcus pneumoniae—Connecticut, 1992-1993. MMWR Morb Mortal Wkly Rep. 1994; 43:216,217,223. http://www.ncbi.nlm.nih.gov/pubmed/8127327?dopt=AbstractPlus

364. Leggiadro RJ. Penicillin- and cephalosporin-resistant Streptococcus pneumoniae: an emerging microbial threat. Pediatrics. 1994; 93:500-3. http://www.ncbi.nlm.nih.gov/pubmed/8115213?dopt=AbstractPlus

365. Bartlett JG. Antibiotic-associated diarrhea. Clin Infect Dis. 1992; 15:573-81. http://www.ncbi.nlm.nih.gov/pubmed/1420669?dopt=AbstractPlus

366. Kelly CP, Pothoulakis C, LaMont JT. Clostridium difficile colitis. N Engl J Med. 1994; 330:257-62. http://www.ncbi.nlm.nih.gov/pubmed/8043060?dopt=AbstractPlus

367. 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; 12:782-4. http://www.ncbi.nlm.nih.gov/pubmed/8414810?dopt=AbstractPlus

368. Centers for Disease Control and Prevention. Drug-resistant Streptococcus pneumoniae—Kentucky and Tennessee, 1993. MMWR Morb Mortal Wkly Rep. 1994; 43:23-7. http://www.ncbi.nlm.nih.gov/pubmed/8277937?dopt=AbstractPlus

369. 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. http://www.ncbi.nlm.nih.gov/pubmed/8451095?dopt=AbstractPlus

370. Chesney PJ. The escalating problem of antimicrobial resistance in Streptococcus pneumoniae . Am J Dis Child. 1992; 146:912-6. http://www.ncbi.nlm.nih.gov/pubmed/1636656?dopt=AbstractPlus

371. Klugman KP. Pneumococcal resistance to antibiotics. Clin Microbiol Rev. 1990; 3:171-96. http://www.ncbi.nlm.nih.gov/pubmed/2187594?dopt=AbstractPlus

372. John CC. Treatment failure with use of a third-generation cephalosporin for penicillin-resistant pneumococcal meningitis: case report and review. Clin Infect Dis. 1994; 18:188-93. http://www.ncbi.nlm.nih.gov/pubmed/8161625?dopt=AbstractPlus

374. Bratzler DW, Dellinger EP, Olsen KM et al. Clinical practice guidelines for antimicrobial prophylaxis in surgery. Am J Health Syst Pharm. 2013; 70:195-283. http://www.ncbi.nlm.nih.gov/pubmed/23327981?dopt=AbstractPlus

375. Gerber MA, Baltimore RS, Eaton CB et al. Prevention of rheumatic fever and diagnosis and treatment of acute Streptococcal pharyngitis: a scientific statement from the American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young, the Interdisciplinary Council on Functional Genomics and Translational Biology, and the Interdisciplinary Council on Quality of Care and Outcomes Research: endorsed by the American Academy of Pediatrics. Circulation. 2009; 119:1541-51. http://www.ncbi.nlm.nih.gov/pubmed/19246689?dopt=AbstractPlus

376. Markowitz M, Gerber MA, Kaplan EL. Treatment of streptococcal pharyngotonsillitis: reports of penicillin’s demise are premature. J Pediatr. 1993; 123:679-85. http://www.ncbi.nlm.nih.gov/pubmed/8229474?dopt=AbstractPlus

377. Pichichero ME, Margolis PA. A comparison of cephalosporins and penicillins in the treatment of group A beta-hemolytic streptococcal pharyngitis: a meta-analysis supporting the concept of microbial copathogenicity. Pediatr Infect Dis J. 1991; 10:275-81. http://www.ncbi.nlm.nih.gov/pubmed/1829514?dopt=AbstractPlus

378. Pichichero ME. Cephalosporins are superior to penicillin for treatment of streptococcal tonsillopharyngitis: is the difference worth it? Pediatr Infect Dis J. 1993; 12:268-74.

379. Stillerman M. Cephalosporins are more effective than penicillin in streptococcal pharyngitis. Pediatr Infect Dis J. 1994; 13:1160. http://www.ncbi.nlm.nih.gov/pubmed/7892097?dopt=AbstractPlus

380. Shulman ST. Cephalosporins are more effective than penicillin in streptococcal pharyngitis. Pediatr Infect Dis J. 1994; 13:1160-1. http://www.ncbi.nlm.nih.gov/pubmed/7892097?dopt=AbstractPlus

381. Milatovic D, Adam D, Hamilton H et al. Cefprozil versus penicillin V in treatment of streptococcal tonsillopharyngitis. Antimicrob Agents Chemother. 1993; 37:1620-3. http://www.ncbi.nlm.nih.gov/pubmed/8215273?dopt=AbstractPlus

382. Aujard Y, Boucot I, Brahimi N et al. Comparative efficacy and safety of four-day cefuroxime axetil and ten-day penicillin treatment of group A beta-hemolytic streptococcal pharyngitis in children. Pediatr Infect Dis J. 1995; 14:295-300. http://www.ncbi.nlm.nih.gov/pubmed/7603811?dopt=AbstractPlus

383. Sanders CC. Cefepime: the next generation? Clin Infect Dis. 1993; 17:369-79.

384. Hardin TC, Jennings TS. Cefepime. Pharmacotherapy. 1994; 14:657-68. http://www.ncbi.nlm.nih.gov/pubmed/7885968?dopt=AbstractPlus

385. Rybak MJ, Palmer SM. Cefepime: part of the new generation. Am J Hosp Pharm. 1994; 51:459-60. http://www.ncbi.nlm.nih.gov/pubmed/8017410?dopt=AbstractPlus

386. Segreti J, Levin S. Bacteriologic and clinical applications of a new extended-spectrum parenteral cephalosporin. Am J Med. 1996; 100(Suppl 6A):45-51S.

387. Barradell LB, Bryson HM. Cefepime. A review of its antibacterial activity, pharmacokinetic properties and therapeutic use. Drugs. 1994; 47:471-505. http://www.ncbi.nlm.nih.gov/pubmed/7514976?dopt=AbstractPlus

388. Hancock REW, Bellido F. Factors involved in the enhanced efficacy against gram-negative bacteria of fourth generation cephalosporins. J Antimicrob Chemother. 1992; 29(Suppl A):1-6. http://www.ncbi.nlm.nih.gov/pubmed/1601751?dopt=AbstractPlus

389. Norrby SR. Side effects of cephalosporins. Drugs. 1987; 34(Suppl 2):105-120. http://www.ncbi.nlm.nih.gov/pubmed/3319495?dopt=AbstractPlus

390. McMahon FG, Ryan JR, Jain AK et al. Absence of disulfiram-type reactions to single and multiple doses of cefonicid: a placebo-controlled study. J Antimicrob Chemother. 1987; 20:913-8. http://www.ncbi.nlm.nih.gov/pubmed/3440774?dopt=AbstractPlus

391. Klass PE, Klein JO. Therapy of bacterial sepsis, meningitis and otitis media in infants and children: 1992 poll of directors of programs in pediatric infectious diseases. Pediatr Infect Dis J. 1992; 11:702-5. http://www.ncbi.nlm.nih.gov/pubmed/1448307?dopt=AbstractPlus

392. Feigin RD, McCracken GH, Klein JO. Diagnosis and management of meningitis. Pediatr Infect Dis J. 1992; 11:785-814. http://www.ncbi.nlm.nih.gov/pubmed/1448332?dopt=AbstractPlus

393. Quagliarello VJ, Scheld WM. Treatment of bacterial meningitis. N Engl J Med. 1997; 336:708-16. http://www.ncbi.nlm.nih.gov/pubmed/9041103?dopt=AbstractPlus

394. Townsend GC, Scheld WM. Infections of the central nervous system. Adv Intern Med. 1998; 43:403-47. http://www.ncbi.nlm.nih.gov/pubmed/9506189?dopt=AbstractPlus

395. Buchingham SC, Brown SP, San Joaquin VH. Breakthrough bacteremia and meningitis during treatment with cephalosporins parenterally for pneumococcal pneumonia. J Pediatr. 1998; 132:174-6. http://www.ncbi.nlm.nih.gov/pubmed/9470026?dopt=AbstractPlus

396. Pacheco TR, Cooper CK, Hardy DJ et al. Failure of cefotaxime treatment in an adult with Streptococcus pneumoniae meningitis. Am J Med. 1997; 102:303-5. http://www.ncbi.nlm.nih.gov/pubmed/9217602?dopt=AbstractPlus

397. Wubbel L, McCracken GH. Management of bacterial meningitis: 1998. Pediatr Rev. 1998; 19:78-84. http://www.ncbi.nlm.nih.gov/pubmed/9509854?dopt=AbstractPlus

398. McIntyre PB, Berkey CS, King SM et al. Dexamethasone as adjunctive therapy in bacterial meningitis: a meta-analysis of randomized clinical trials since 1988. JAMA. 1997; 278:925-31. http://www.ncbi.nlm.nih.gov/pubmed/9302246?dopt=AbstractPlus

399. Cabellos C, Viladrich PF, Verdaguer R et al. A single daily dose of ceftriaxone for bacterial meningitis in adults: experience with 84 patients and review of the literature. Clin Infect Dis. 1995; 20:1164-8. http://www.ncbi.nlm.nih.gov/pubmed/7619994?dopt=AbstractPlus

400. Chesney PJ, Halsey NA, Marcy SM. Treatment of bacterial meningitis. N Engl J Med. 1997; 337:793-4. http://www.ncbi.nlm.nih.gov/pubmed/9289652?dopt=AbstractPlus

401. Quagliarello V, Scheld WM. Treatment of bacterial meningitis. N Engl J Med. 1997; 337:794. http://www.ncbi.nlm.nih.gov/pubmed/9289653?dopt=AbstractPlus

402. Bhattacharya S. Management of meningitis caused by penicillin-resistant Streptococcus pneumoniae . Antimicrob Agents Chemother. 1996; 40:827-8. http://www.ncbi.nlm.nih.gov/pubmed/8851626?dopt=AbstractPlus

403. Ahmed A. A critical evaluation of vancomycin for treatment of bacterial meningitis. Pediatr Infect Dis J. 1997; 16:895-903. http://www.ncbi.nlm.nih.gov/pubmed/9306486?dopt=AbstractPlus

404. Mathisen GE, Johnson JP. Brain abscess. Clin Infect Dis. 1997; 25:763-81. http://www.ncbi.nlm.nih.gov/pubmed/9356788?dopt=AbstractPlus

405. American Academy of Pediatrics Committee on Infectious Diseases. Therapy for children with invasive pneumococcal infections. Pediatrics. 1997; 99:289-99. http://www.ncbi.nlm.nih.gov/pubmed/9024464?dopt=AbstractPlus

406. Reviewers’ comments (personal observations).

407. Leggiadro RJ. Penicillin- and cephalosporin-resistant Streptococcus pneumoniae: an emerging threat. Pediatrics. 1994;93:500-3. http://www.ncbi.nlm.nih.gov/pubmed/8115213?dopt=AbstractPlus

408. Centers for Disease Control and Prevention. Drug-resistant Streptococcus pneumoniae—Kentucky and Tennessee, 1993. MMWR Morb Mortal Wkly Rep. 1994; 43:23-7. http://www.ncbi.nlm.nih.gov/pubmed/8277937?dopt=AbstractPlus

409. John CC. Treatment failure with use of a third-generation cephalosporin for penicillin-resistant pneumococcal meningitis: case report and review. Clin Infect Dis. 1994; 18:188-93. http://www.ncbi.nlm.nih.gov/pubmed/8161625?dopt=AbstractPlus

410. Pallares R, Linares J, Vadillo M et al. Resistance to penicillin and cephalosporin and mortality from severe pneumococcal pneumonia in Barcelona, Spain. N Engl J Med. 1995; 333:474-80. http://www.ncbi.nlm.nih.gov/pubmed/7623879?dopt=AbstractPlus

411. Hofmann J, Cetron MS, Farley MM et al. The prevalence of drug-resistant Streptococcus pneumoniae in Atlanta. N Engl J Med. 1995; 333:481-6. http://www.ncbi.nlm.nih.gov/pubmed/7623880?dopt=AbstractPlus

412. Panel on Opportunistic Infections in HIV-infected Adults and Adolescents. Guidelines for prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America (May 7, 2013). Updates may be available at HHS AIDS Information (AIDSinfo) website. http://www.aidsinfo.nih.gov

413. 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. http://www.ncbi.nlm.nih.gov/pubmed/8451095?dopt=AbstractPlus

414. Lorber B. Listeriosis. Clin Infect Dis. 1997; 24:1-11. http://www.ncbi.nlm.nih.gov/pubmed/8994747?dopt=AbstractPlus

415. Mathisen GE, Johnson JP. Brain abscess. Clin Infect Dis. 1997; 25:763-81. http://www.ncbi.nlm.nih.gov/pubmed/9356788?dopt=AbstractPlus

416. Tunkel AR, Scheld WM. Issues in the management of bacterial meningitis. Am Fam Physician. 1997; 56:1355-62. http://www.ncbi.nlm.nih.gov/pubmed/9337758?dopt=AbstractPlus

417. Cunha BA. Treatment of pelvic inflammatory disease. Clin Pharm. 1990; 9:275-85. http://www.ncbi.nlm.nih.gov/pubmed/2184973?dopt=AbstractPlus

418. Walker CK, Kahn JG, Washington AE et al. Pelvic inflammatory disease: metaanalysis of antimicrobial regimen efficacy. J Infect Dis. 1993; 168:969-78. http://www.ncbi.nlm.nih.gov/pubmed/8376843?dopt=AbstractPlus

419. Hemsell DL, Little BB, Faro S et al. Comparison of three regimens recommended by the Centers for Disease Control and Prevention for the treatment of women hospitalized with acute pelvic inflammatory disease. Clin Infect Dis. 1994; 19:720-7. http://www.ncbi.nlm.nih.gov/pubmed/7803638?dopt=AbstractPlus

420. 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; 24:170-8. http://www.ncbi.nlm.nih.gov/pubmed/9114143?dopt=AbstractPlus

421. Green SM, Rothrock SG. Single-dose intramuscular ceftriaxone for acute otitis media in children. Pediatrics. 1993; 91:23-30. http://www.ncbi.nlm.nih.gov/pubmed/8416502?dopt=AbstractPlus

422. Varsano I, Frydman M, Amir J et al. Single intramuscular dose of ceftriaxone as compared to 7-day amoxicillin therapy for acute otitis media in children: a double-blind clinical trial. Chemotherapy. 1988; 34(Suppl 1):39-46. http://www.ncbi.nlm.nih.gov/pubmed/3246170?dopt=AbstractPlus

423. Bauchner H, Adams W, Barnett E et al. Therapy for acute otitis media: preference of parents for oral or parenteral antibiotics. Arch Pediatr Adolesc Med. 1996; 150:396-9. http://www.ncbi.nlm.nih.gov/pubmed/8634735?dopt=AbstractPlus

424. Goldfarb J, Medendorp S. New therapies for otitis media. Clin Pediatr. 1994; Nov:647-8.

425. Reynolds RD. Cefaclor and serum sickness-like reaction. JAMA. 1996; 276:950. http://www.ncbi.nlm.nih.gov/pubmed/8805721?dopt=AbstractPlus

426. Kelsey DK. Cefaclor and serum sickness-like reaction. JAMA. 1996; 276:950-1. http://www.ncbi.nlm.nih.gov/pubmed/8805721?dopt=AbstractPlus

427. Stricker BH, Tussen JGP. Serum sickness-like reactions to cefaclor. J Clin Epidemiol. 1992; 45:1177-84. http://www.ncbi.nlm.nih.gov/pubmed/1474414?dopt=AbstractPlus

428. Platt R, Dreis MW, Kennedy DL et al. Serum sickness-like reactions to amoxicillin, cefaclor, cephalexin, and trimethoprim-sulfamethoxazole. J Infect Dis. 1988; 158:474-7. http://www.ncbi.nlm.nih.gov/pubmed/3261315?dopt=AbstractPlus

429. Heckbert SR, Stryker WS, Coltin KL et al. Serum sickness in children after antibiotic exposure: estimates of occurrence and morbidity in a health maintenance organization population. Am J Epidemiol. 1990; 132:336-42. http://www.ncbi.nlm.nih.gov/pubmed/2115293?dopt=AbstractPlus

430. Hebert AA, Sigman ES, Levy ML. Serum sickness-like reactions from cefaclor in children. J Am Acad Dermatol. 1991; 25:805-8. http://www.ncbi.nlm.nih.gov/pubmed/1802903?dopt=AbstractPlus

431. Vial T, Pont J, Pham E et al. Cefaclor-associated serum sickness-like disease: eight cases and review of the literature. Ann Pharmacother. 1992; 26:910-4. http://www.ncbi.nlm.nih.gov/pubmed/1504397?dopt=AbstractPlus

432. Lowry N, Kearns GL, Young RA et al. Serum sickness-like reactions associated with cefprozil therapy. J Pediatr. 1994; 125:325-8. http://www.ncbi.nlm.nih.gov/pubmed/8040786?dopt=AbstractPlus

433. Klein JO. Selection of oral antimicrobial agents for otitis media and pharyngitis. Infect Dis Clin Pract. 1995; 4(Suppl 2):S88-94.

434. 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; 12:782-4. http://www.ncbi.nlm.nih.gov/pubmed/8414810?dopt=AbstractPlus

435. Klein JO. Management of streptococcal pharyngitis. Pediatr Infect Dis J. 1994; 13:572-5. http://www.ncbi.nlm.nih.gov/pubmed/8078757?dopt=AbstractPlus

436. Pichichero ME, Cohen R. Shortened course of antibiotic therapy for acute otitis media, sinusitis and tonsillopharyngitis. Pediatr Infect Dis J. 1997; 16:680-95. http://www.ncbi.nlm.nih.gov/pubmed/9239773?dopt=AbstractPlus

437. Dajani AS, Kessler SL, Mendelson R et al. Cefpodoxime proxetil vs. penicillin V in pediatric streptococcal pharyngitis/tonsillitis. Pediatr Infect Dis J. 1993; 12:275-9. http://www.ncbi.nlm.nih.gov/pubmed/8483620?dopt=AbstractPlus

438. Tack KJ, Henry DC, Gooch WM et al et al. Five-day cefdinir treatment for streptococcal pharyngitis. Antimicrob Agents Chemother. 1998; 42:1073-5. http://www.ncbi.nlm.nih.gov/pubmed/9593129?dopt=AbstractPlus

439. Mehra S, Van Moerkerke M, Welck J et al. Short course therapy with cefuroxime axetil for group A streptococcal tonsillopharyngitis in children. Pediatr Infect Dis J. 1998; 17:452-7. http://www.ncbi.nlm.nih.gov/pubmed/9655533?dopt=AbstractPlus

440. Milatovic D. Evaluation of cefadroxil, penicillin and erythromycin in the treatment of streptococcal tonsillopharyngitis. Pediatr Infect Dis J. 1991; 10:S61-3. http://www.ncbi.nlm.nih.gov/pubmed/1945599?dopt=AbstractPlus

441. Cohen R. Clinical experience with cefpodoxime proxetil in acute otitis media. Pediatr Infect Dis J. 1995; 14:S12-8. http://www.ncbi.nlm.nih.gov/pubmed/7792125?dopt=AbstractPlus

442. Fernandez GJ, MacLoughlin GJF, Barreto DG et al. Cefpodoxime proxetil suspension compared with cefaclor suspension for treatment of acute otitis media in paediatric patients. J Antimicrob Chemother. 1996; 37:565-73. http://www.ncbi.nlm.nih.gov/pubmed/9182113?dopt=AbstractPlus

443. Asmar BI, Dajani AS, Del Beccaro MA et al. Comparison of cefpodoxime proxetil and cefixime in the treatment of acute otitis media in infants and children. Pediatrics. 1994; 94:847-52. http://www.ncbi.nlm.nih.gov/pubmed/7971000?dopt=AbstractPlus

444. Kozyrskyj AL, Hildes-Ripstein GE, Longstaffe SEA et al. Treatment of acute otitis media with a shortened course of antibiotics: a meta-analysis. JAMA. 1998; 279:1736-42. http://www.ncbi.nlm.nih.gov/pubmed/9624028?dopt=AbstractPlus

445. Pichichero ME, McLinn S, Aronovitz G et al. Cefprozil treatment of persistent and recurrent acute otitis media. Pediatr Infect Dis J. 1997; 16:471-8. http://www.ncbi.nlm.nih.gov/pubmed/9154539?dopt=AbstractPlus

446. Hendrickse WA, Kusmiesz H, Shelton S et al. Five-day vs. ten days of therapy for acute otitis media. Pediatr Infect Dis J. 1988; 7:14-23. http://www.ncbi.nlm.nih.gov/pubmed/3277153?dopt=AbstractPlus

447. Kafetzis DA, Astra H, Mitropoulos L. Five-day versus ten-day treatment of acute otitis media with cefprozil. Eur J Clin Microbiol Infect Dis. 1997; 16:283-6. http://www.ncbi.nlm.nih.gov/pubmed/9177961?dopt=AbstractPlus

448. McCracken GH. Treatment of acute otitis media in an era of increasing microbial resistance. Pediatr Infect Dis J. 1998; 17:576-9. http://www.ncbi.nlm.nih.gov/pubmed/9655564?dopt=AbstractPlus

449. Klein JO. Otitis Media. Clin Infect Dis J. 1994; 19:823-33.

450. Baddour LM, Wilson WR, Bayer AS. Infective endocarditis: diagnosis, antimicrobial therapy, and management of complications: a statement for healthcare professionals from the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease of the Young, and the Councils on Clinical Cardiology, Stroke, and Cardiovascular Surgery and Anesthesia, American Heart Association: endorsed by the Infectious Diseases Society of. Circulation. 2005; 111: e394-433. http://www.ncbi.nlm.nih.gov/pubmed/15956145?dopt=AbstractPlus

451. Wilson W, Taubert KA, Gewitz M et al. Prevention of infective endocarditis: guidelines from the American Heart Association: a guideline from the American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee, Council on Cardiovascular Disease in the Young, and the Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and the Quality of Care and Outcomes Research Interdisciplinary Working Group. Circulation. 2007; 116:1736-54. http://www.ncbi.nlm.nih.gov/pubmed/17446442?dopt=AbstractPlus

453. Adu A, Armour CL. Drug utilisation review (DUR) of the third generation cephalosporins: focus on ceftriaxone, ceftazidime and cefotaxime. Drugs. 1995; 50:423-39. http://www.ncbi.nlm.nih.gov/pubmed/8521766?dopt=AbstractPlus

454. Charnas R, Luthi AR, Ruch W et al. Once daily ceftriaxone plus amikacin vs. three times daily ceftazidime plus amikacin for treatment of febrile neutropenic children with cancer. Pediatr Infect Dis J. 1997; 16:346-53. http://www.ncbi.nlm.nih.gov/pubmed/9109134?dopt=AbstractPlus

455. 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; 119:584-93. http://www.ncbi.nlm.nih.gov/pubmed/8363169?dopt=AbstractPlus

456. Cordonnier C, Herbrecht R, Pico JL et al et al. cefepime/amikacin versus ceftazidime/amikacin as empirical therapy for febrile episodes in neutropenic patients: a comparative study. Clin Infect Dis. 1997; 24:41-51. http://www.ncbi.nlm.nih.gov/pubmed/8994754?dopt=AbstractPlus

457. Freifeld AG, Bow EJ, Sepkowitz KA et al. Clinical Practice Guideline for the Use of Antimicrobial Agents in Neutropenic Patients with Cancer: 2010 Update by the Infectious Disease Society of America. Clin Infect Dis. 2011; 52:e56-93.

458. Pizzo PA. Management of fever in patients with cancer and treatment-induced neutropenia. N Engl J Med. 1993; 328:1323-32. http://www.ncbi.nlm.nih.gov/pubmed/8469254?dopt=AbstractPlus

459. Ramphal R, Gucalp R, Rotstein C et al. Clinical experience with single agent and combination regimens in the management of infection in the febrile neutropenic patient. Am J Med. 1996; 100(Suppl 6A):83S-89S. http://www.ncbi.nlm.nih.gov/pubmed/8678102?dopt=AbstractPlus

460. Viscoli C. The evolution of the empirical management of fever and neutropenia in cancer patients. J Antimicrob Chemother. 1998; 41(Suppl D):65-80. http://www.ncbi.nlm.nih.gov/pubmed/9688453?dopt=AbstractPlus

461. Rolston KV. Expanding the options for risk-based therapy in febrile neutropenia. Diagn Microbiol Infect Dis. 1998; 31:411-6. http://www.ncbi.nlm.nih.gov/pubmed/9635917?dopt=AbstractPlus

462. Link H, Maschmeyer G, Meyer P et al. Interventional antimicrobial therapy in febrile neutropenic patients. Ann Hematol. 1994; 69:231-43. http://www.ncbi.nlm.nih.gov/pubmed/7948312?dopt=AbstractPlus

463. Giamarellou H. Empiric therapy for infections in the febrile, neutropenic, compromised host. Med Clin N Am. 1995; 79:559-77. http://www.ncbi.nlm.nih.gov/pubmed/7752729?dopt=AbstractPlus

464. Karthaus M, Wolf HH, Kampfe D et al. Ceftriaxone monotherapy in the treatment of low-risk febrile neutropenia. Chemotherapy. 1998; 44:343-54. http://www.ncbi.nlm.nih.gov/pubmed/9732151?dopt=AbstractPlus

465. Bartolozzi S, Clerico A, Properzi E et al. Ceftriaxone as a single agent in empirical therapy of unexplained fever in granulocytopenic children with solid tumors. J Chemother. 1997; 9:227-31. http://www.ncbi.nlm.nih.gov/pubmed/9210007?dopt=AbstractPlus

466. Eggimann P, Glauser MP, Aoun M et al. Cefepime monotherapy for the empirical treatment of fever in granulocytopenic cancer patients. J Antimicrob Chemother. 1993; 32(Suppl B):151-63. http://www.ncbi.nlm.nih.gov/pubmed/8150759?dopt=AbstractPlus

467. Yamamura D, Gucalp R, Carlisle P et al. Open randomized study of cefepime versus piperacillin-gentamicin for treatment of febrile neutropenic cancer patients. Antimicrob Agents Chemother. 1997; 41:1704-8. http://www.ncbi.nlm.nih.gov/pubmed/9257745?dopt=AbstractPlus

468. Hathorn J, Chandrasakar P, Baird I et al. Double-blind comparison of cefepime vs ceftazidime to treat febrile neutropenia. Proceedings of ICAAC New Orleans 1996. Abstract No. LM22.

469. Biron P, Fuhrmann C, Kheder R et al. Cefepime (CEP) vs imipenem-cilastatin (IMP) as empiric monotherapy in 400 febrile patients with short duration neutropenia. Proceedings of ICAAC Toronto 1997. Abstract No. LM48.

470. Anon. Treatment of Lyme disease. Med Lett Drugs Ther. 2000; 42:37-9. http://www.ncbi.nlm.nih.gov/pubmed/10825919?dopt=AbstractPlus

471. Steere AC. Lyme disease. N Engl J Med. 1989; 321:586-96. http://www.ncbi.nlm.nih.gov/pubmed/2668764?dopt=AbstractPlus

472. Luft BJ, Gorevic PD, Halperin JJ et al. A perspective on the treatment of Lyme borreliosis. Rev Infect Dis. 1989; 11(Suppl 6):S1518-25.

473. Neu HC. A perspective on therapy of Lyme infection. Ann NY Acad Sci. 1988; 539:314-6. http://www.ncbi.nlm.nih.gov/pubmed/3056200?dopt=AbstractPlus

474. Reviewers’ comments on Lyme disease revisions (personal observations) on ceftriaxone 8:12.06.12.

475. Centers for Disease Control and Prevention. Ceftriaxone-associated biliary complications of treatment of suspected disseminated Lyme disease—New Jersey, 1990-1992. MMWR Morb Mortal Wkly Rep. 1993; 42:39-42. http://www.ncbi.nlm.nih.gov/pubmed/8419791?dopt=AbstractPlus

476. Spach DH, Liles WC, Campbell GL et al. Tick-borne diseases in the United States. N Engl J Med. 1993; 329:936-47. http://www.ncbi.nlm.nih.gov/pubmed/8361509?dopt=AbstractPlus

477. Luft BJ, Gardner P, Lightfoot RW Jr. Appropriateness of parenteral antibiotic treatment for patients with presumed Lyme disease. Ann Intern Med. 1993; 119:518. http://www.ncbi.nlm.nih.gov/pubmed/8357119?dopt=AbstractPlus

478. Lightfoot RW, Luft BJ, Rahn DW et al. Empiric parenteral antibiotic treatment of patients with fibromyalgia and fatigue and a positive serologic result for Lyme disease. A cost-effectiveness analysis. Ann Intern Med. 1993; 119:503-9. http://www.ncbi.nlm.nih.gov/pubmed/8357117?dopt=AbstractPlus

479. Bakken LL, Case KL, Callister SM et al. Performance of 45 laboratories participating in a proficiency testing program for Lyme disease serology. JAMA. 1992; 268:891-5. http://www.ncbi.nlm.nih.gov/pubmed/1640618?dopt=AbstractPlus

480. Nocton JJ, Steere AC. Lyme disease. Adv Intern Med. 1995; 40:69-117. http://www.ncbi.nlm.nih.gov/pubmed/7747659?dopt=AbstractPlus

481. Jantausch BA. Lyme disease, Rocky Mountain spotted fever, ehrlichiosis: emerging and established challenges for the clinician. Ann Allergy. 1994; 73:4-11. http://www.ncbi.nlm.nih.gov/pubmed/8030801?dopt=AbstractPlus

482. Nadelman RB, Wormser GP. Erythema migrans and early Lyme disease. Am J Med. 1995; 98(4A):15-23S.

483. Sigal LH. Early disseminated Lyme disease: cardiac manifestations. Am J Med. 1995; 98(4A):25-28S.

484. Steere AC. Musculoskeletal manifestations of Lyme disease. Am J Med. 1995; 98(4A):44-48S.

485. Sigal LH. Management of Lyme disease refractory to antibiotic therapy. Rheum Dis Clin North Am. 1995; 21:217-30. http://www.ncbi.nlm.nih.gov/pubmed/7732170?dopt=AbstractPlus

486. Dattwyler RJ, Luft BJ, Kunkel MJ et al. Ceftriaxone compared with doxycycline for the treatment of acute disseminated lyme disease. N Engl J Med. 1997; 337:289-94. http://www.ncbi.nlm.nih.gov/pubmed/9233865?dopt=AbstractPlus

487. Shapiro ED. Lyme disease in children. Am J Med. 1995; 98(4A):69-73S.

488. Pachner AR. Early disseminated Lyme disease: Lyme meningitis. Am J Med. 1995; 98(4A):30-37S.

489. Sigal LH. Persisting symptoms of Lyme disease—possible explanations and implications for treatment. J Rheumatol. 1994; 21:593-5. http://www.ncbi.nlm.nih.gov/pubmed/8035381?dopt=AbstractPlus

490. Rees DHE, Axford JS. Lyme arthritis. Ann Rheum Dis. 1994; 53:553-6. http://www.ncbi.nlm.nih.gov/pubmed/7979590?dopt=AbstractPlus

491. Ettestad PJ, Campbell GL, Welbel SF et al. Biliary complications in the treatment of unsubstantiated Lyme disease. J Infect Dis. 1995; 171:356-61. http://www.ncbi.nlm.nih.gov/pubmed/7844372?dopt=AbstractPlus

492. Hassler D, Zoller L, Haude M et al. Cefotaxime versus penicillin in the late stage of Lyme disease—prospective, randomized therapeutic study. Infection. 1990; 18:16-20. http://www.ncbi.nlm.nih.gov/pubmed/2179134?dopt=AbstractPlus

493. Pfister HW, Preac-Mursic V, Wilske B et al. Cefotaxime vs penicillin G for acute neurologic manifestations in Lyme borreliosis. Arch Neurol. 1989; 46:1190-4. http://www.ncbi.nlm.nih.gov/pubmed/2684107?dopt=AbstractPlus

494. Steere AC, Grodzicki RL, Kornblatt AN et al. The spirochetal etiology of Lyme disease. N Engl J Med. 1983; 308:740-2. http://www.ncbi.nlm.nih.gov/pubmed/6828119?dopt=AbstractPlus

495. Meyerhoff J. Lyme disease. Am J Med. 1983; 75:663-70. http://www.ncbi.nlm.nih.gov/pubmed/6353917?dopt=AbstractPlus

496. Steere AC, Malawista SE, Newman JH et al. Antibiotic therapy in Lyme disease. Ann Intern Med. 1980; 93:1-8. http://www.ncbi.nlm.nih.gov/pubmed/6967272?dopt=AbstractPlus

497. Benach JL, Boster EM, Hanrahan JP et al. Spirochetes isolated from the blood of two patients with Lyme disease. N Engl J Med. 1983; 308:740-2. http://www.ncbi.nlm.nih.gov/pubmed/6828119?dopt=AbstractPlus

498. Steere AC, Hutchinson GJ, Rahn DW et al. Treatment of early manifestations of Lyme disease. Ann Intern Med. 1983; 99:22-6. http://www.ncbi.nlm.nih.gov/pubmed/6407378?dopt=AbstractPlus

499. Centers for Disease Control. Update: Lyme disease—United States. MMWR Morb Mortal Wkly Rep. 1984; 33:268-70. http://www.ncbi.nlm.nih.gov/pubmed/6425627?dopt=AbstractPlus

500. Centers for Disease Control. Penicillin therapy in Lyme disease. MMWR Morb Mortal Wkly Rep. 1980; 29:237-9.

501. Steere AC, Green J, Schoen RT et al. Successful parenteral penicillin therapy of established lyme arthritis. N Engl J Med. 1985; 312:869-74. http://www.ncbi.nlm.nih.gov/pubmed/3883177?dopt=AbstractPlus

502. Centers for Disease Control. Update: Lyme disease and cases occurring during pregnancy—United States. MMWR Morb Mortal Wkly Rep. 1985; 34:376-8,383-4. http://www.ncbi.nlm.nih.gov/pubmed/3925314?dopt=AbstractPlus

503. Centers for Disease Control and Prevention. Diagnosis and management of foodborne illness. A primer for physicians. MMWR Recomm Rep. 2001; 50(RR-2):1-69. http://www.cdc.gov/mmwr/PDF/rr/rr5002.pdf

504. Dattwyler RJ, Halperin JJ, Volkman DJ et al. Treatment of late Lyme borreliosis—randomized comparison of ceftriaxone and penicillin. Lancet. 1988; 1:1191-4. http://www.ncbi.nlm.nih.gov/pubmed/2897008?dopt=AbstractPlus

505. Eichenfield AH, Athreya BH. Lyme disease: of ticks and titers. J Pediatr. 1989; 114:328-33. http://www.ncbi.nlm.nih.gov/pubmed/2644410?dopt=AbstractPlus

506. Ciesielski CA, Markowitz LE, Horsley R et al. Lyme disease surveillance in the United States, 1983—1986. Rev Infect Dis. 1989; 11(Suppl 6):S1435-41. http://www.ncbi.nlm.nih.gov/pubmed/2682955?dopt=AbstractPlus

507. Centers for Disease Control. Lyme disease—Connecticut. MMWR Morb Mortal Wkly Rep. 1988; 37:1-3. http://www.ncbi.nlm.nih.gov/pubmed/3122001?dopt=AbstractPlus

508. Asbrink E, Olsson I. Clinical manifestations of erythema chronicum migrans Afzelius in 161 patients. A comparison with Lyme disease. Acta Derm Venereol (Stockh). 1985; 65:43-52.

509. Luft BJ, Volkman DJ, Halperin JJ et al. New chemotherapeutic approaches in the treatment of Lyme borreliosis. Ann NY Acad Sci. 1988; 539:352-61. http://www.ncbi.nlm.nih.gov/pubmed/3056203?dopt=AbstractPlus

510. Mursic VP, Wilske B, Schierz G. In vitro and in vivo susceptibility of Borrelia burgdorferi . Eur J Clin Microbiol. 1987; 6:424-6. http://www.ncbi.nlm.nih.gov/pubmed/3665899?dopt=AbstractPlus

511. Johnson RC, Kodner C, Russell M. In vitro and in vivo susceptibility of the Lyme disease spirochete, Borrelia burgdorferi, to four antimicrobial agents. Antimicrob Agents Chemother. 1987; 31:164-7. http://www.ncbi.nlm.nih.gov/pubmed/3566246?dopt=AbstractPlus

512. Dattwyler RJ, Halperin JJ, Pass H et al. Ceftriaxone as effective therapy in refractory Lyme disease. J Infect Dis. 1987; 155:1322-5. http://www.ncbi.nlm.nih.gov/pubmed/3572042?dopt=AbstractPlus

513. Rahn DW, Malawista SE. Lyme disease: recommendations for diagnosis and treatment. Ann Intern Med. 1991; 114:472-81. http://www.ncbi.nlm.nih.gov/pubmed/1994795?dopt=AbstractPlus

514. Nadelman RB, Wormser GP. Lyme borreliosis. Lancet. 1998; 352:557-65. http://www.ncbi.nlm.nih.gov/pubmed/9716075?dopt=AbstractPlus

515. Eckman MH, Steere AC, Kalish RA et al. Cost effectiveness of oral as compared with intravenous antibiotic treatment for patients with early Lyme disease or Lyme arthritis. N Engl J Med. 1997; 337:357-63. http://www.ncbi.nlm.nih.gov/pubmed/9233874?dopt=AbstractPlus

516. Rahn DW, Felz MW. Lyme disease update. Current approach to early, disseminated, and late disease. Postgrad Med. 1998;103:51-4, 57-9, 63-4. http://www.ncbi.nlm.nih.gov/pubmed/9590986?dopt=AbstractPlus

517. Collins JJ. Treatment of erythema migrans. Ann Intern Med. 1997; 126:408. http://www.ncbi.nlm.nih.gov/pubmed/9054288?dopt=AbstractPlus

518. Nadelman RB, Luger SW, Frank E et al. Comparison of cefuroxime axetil and doxycycline in the treatment of early Lyme disease. Ann Intern Med. 1992; 117:273-280. http://www.ncbi.nlm.nih.gov/pubmed/1637021?dopt=AbstractPlus

519. Luger SW, Paparone P, Wormser GP et al. Comparison of cefuroxime axetil and doxycycline in treatment of patients with early Lyme disease associated with erythema migrans. Antimicrob Agents Chemother. 1995; 39:661-7. http://www.ncbi.nlm.nih.gov/pubmed/7793869?dopt=AbstractPlus

520. Perry CM, Brogden RN. Cefuroxime axetil: a review of its antibacterial activity, pharmacokinetic properties and therapeutic efficacy. Drugs. 1996; 52:125-58. http://www.ncbi.nlm.nih.gov/pubmed/8799689?dopt=AbstractPlus

521. Glaxo Wellcome Inc. Ceftin tablets: use in Lyme disease. Research Triangle Park, NC (undated).

522. 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 Streptococcal pneumoniae Therapeutic Working Group. Pediatr Infect Dis J. 1999; 18:1-9. http://www.ncbi.nlm.nih.gov/pubmed/9951971?dopt=AbstractPlus

523. Dagan R, Leibovitz E, Greenberg D et al. Early eradication of pathogens from middle ear fluid during antibiotic treatment of acute otitis media is associated with improved clinical outcome. Pediatr Infect Dis J. 1998; 17:776-82. http://www.ncbi.nlm.nih.gov/pubmed/9779760?dopt=AbstractPlus

524. Klein JO. Current recommendations on the therapy of otitis media. Pediatr Infect Dis J. 1998; 17:1058-9. http://www.ncbi.nlm.nih.gov/pubmed/9849999?dopt=AbstractPlus

525. Gehanno P, N’Guyen L, Derriennic M et al. Pathogens isolated during treatment failures in otitis. Pediatr Infect Dis J. 1998; 17:885-90. http://www.ncbi.nlm.nih.gov/pubmed/9802629?dopt=AbstractPlus

526. Bluestone CD. Role of surgery for otitis media in the era of resistant bacteria. Pediatr Infect Dis J. 1998; 17:1090-8. http://www.ncbi.nlm.nih.gov/pubmed/9850004?dopt=AbstractPlus

527. Klein JO. Clinical implications of antibiotic resistance for management of acute otitis media. Pediatr Infect Dis J. 1998; 17:1084-9. http://www.ncbi.nlm.nih.gov/pubmed/9850003?dopt=AbstractPlus

528. Blumer JL. Pharmacokinetics and pharmacodynamics of new and old antimicrobial agents for acute otitis media. Pediatr Infect Dis J. 1998; 17:1070-5. http://www.ncbi.nlm.nih.gov/pubmed/9850001?dopt=AbstractPlus

529. 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. http://www.ncbi.nlm.nih.gov/pubmed/9655535?dopt=AbstractPlus

530. Zeisel SA, Roberts JE, Neebe EC et al. A longitudinal study of otitis media with effusion among 2- to 5-year-old African-American children in child care. Pediatrics. 1999; 103:15-9. http://www.ncbi.nlm.nih.gov/pubmed/9917433?dopt=AbstractPlus

531. Blumer JL. Implications of pharmacokinetics in making choices for the management of acute otitis media. Pediatr Infect Dis J. 1998; 17:565-70. http://www.ncbi.nlm.nih.gov/pubmed/9655562?dopt=AbstractPlus

532. Klein JO. Protecting the therapeutic advantage of antimicrobial agents used for otitis media. Pediatr Infect Dis J. 1998; 17:571-5. http://www.ncbi.nlm.nih.gov/pubmed/9655563?dopt=AbstractPlus

533. Heikkinen T, Thint M, Chonmaitree T. Prevalence of various respiratory viruses in the middle ear during acute otitis media. N Engl J Med. 1999; 340:260-4. http://www.ncbi.nlm.nih.gov/pubmed/9920949?dopt=AbstractPlus

534. Eskola J, Hovi T. Respiratory viruses in acute otitis media. N Engl J Med. 1999; 340:312-4. http://www.ncbi.nlm.nih.gov/pubmed/9920959?dopt=AbstractPlus

535. 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; 43:16-20. http://www.ncbi.nlm.nih.gov/pubmed/9869558?dopt=AbstractPlus

536. Hueston WJ, Ornstein S, Jenkins RG et al. Treatment of recurrent otitis media after a previous treatment failure: which antibiotics work best? J Fam Pract. 1999; 48:43-6.

537. Jacobs MR. Antibiotic-resistant Streptococcus pneumoniae in acute otitis media: overview and update. Pediatr Infect Dis J. 1998; 17:947-52. http://www.ncbi.nlm.nih.gov/pubmed/9802651?dopt=AbstractPlus

538. Poole MD. Implications of drug-resistant Streptococcus pneumoniae for otitis media. Pediatr Infect Dis J. 1998; 17:953-6. http://www.ncbi.nlm.nih.gov/pubmed/9802652?dopt=AbstractPlus

539. Harkness MJ. Controversies: treatment of acute otitis media. JAMA. 1998; 279:1783. http://www.ncbi.nlm.nih.gov/pubmed/9628701?dopt=AbstractPlus

540. Pichichero ME. Changing the treatment paradigm for acute otitis media in children. JAMA. 1998; 279:1748-50. http://www.ncbi.nlm.nih.gov/pubmed/9624032?dopt=AbstractPlus

541. Hirschmann JV. Methods for decreasing antibiotic use in otitis media. Lancet. 1998; 352:672. http://www.ncbi.nlm.nih.gov/pubmed/9728980?dopt=AbstractPlus

542. Cohen R, Levy C, Boucherat M et al. A multicenter, randomized, double-blind trial of 5 versus 10 days of antibiotic therapy for acute otitis media in young children. J Pediatr. 1998; 133:634-9. http://www.ncbi.nlm.nih.gov/pubmed/9821420?dopt=AbstractPlus

543. Dowell SF, Marcy SM, Phillips WR et al. Otitis media—principles of judicious use of antimicrobial agents. Pediatrics. 1998; 101:165-71.

544. Pichichero ME, Pichichero Cl. Persistent acute otitis media: I. Causative pathogens. Pediatr Infect Dis J. 1995; 14:178-83. http://www.ncbi.nlm.nih.gov/pubmed/7761181?dopt=AbstractPlus

545. Pichichero ME, Pichichero CL. Persistent acute otitis media: II. Antimicrobial treatment. Pediatr Infect Dis J. 1995; 13:183-8.

546. Paradise JL. Managing otitis media: a time for change. Pediatrics. 1995; 96:712-5. http://www.ncbi.nlm.nih.gov/pubmed/7567336?dopt=AbstractPlus

547. Williams RL, Chalmers TC, Stange KC et al. Use of antibiotics in preventing recurrent acute otitis media and in treating otitis media with effusion: a meta-analytic attempt to resolve the brouhaha. JAMA. 1993; 270:1344-51. http://www.ncbi.nlm.nih.gov/pubmed/8141875?dopt=AbstractPlus

548. Rosenfeld RM, Vertrees JE, Carr J et al. Clinical efficacy of antimicrobial drugs for acute otitis media: metaanalysis of 5400 children from thirty-three randomized trials. J Pediatr. 1994; 124:355-67. http://www.ncbi.nlm.nih.gov/pubmed/8120703?dopt=AbstractPlus

549. Hamrick HJ, Garfunkel JM. Therapy for acute otitis media: applicability of metaanalysis to the individual patient. J Pediatr. 1994; 124:431. http://www.ncbi.nlm.nih.gov/pubmed/8120714?dopt=AbstractPlus

550. Rosenfeld RM. Controversies: treatment of acute otitis media. JAMA. 1998; 279:1784. http://www.ncbi.nlm.nih.gov/pubmed/9628703?dopt=AbstractPlus

551. Faden H, Doern G, Wolf J et al. Antimicrobial susceptibility of nasopharyngeal isolates of potential pathogens recovered from infants before antibiotic therapy: implications for the management of otitis media. Pediatr Infect Dis J. 1994; 13:609-12. http://www.ncbi.nlm.nih.gov/pubmed/7970948?dopt=AbstractPlus

552. Rodriguez WJ, Schwartz RH, Thorne MM. Increasing incidence of penicillin- and ampicillin-resistant middle ear pathogens. Pediatr Infect Dis J. 1995; 14:1075-8. http://www.ncbi.nlm.nih.gov/pubmed/8745021?dopt=AbstractPlus

553. Cohen R, Bingen E, Varon E et al. Change in nasopharyngeal carriage of Streptococcus pneumoniae resulting from antibiotic therapy for acute otitis media in children. Pediatr Infect Dis J. 1997; 16:555-60. http://www.ncbi.nlm.nih.gov/pubmed/9194104?dopt=AbstractPlus

554. Indudharan R, Haq JA, Aiyar S. Antibiotics in chronic suppurative otitis media: a bacteriologic study. Ann Otol Rhinol Laryngol. 1999; 108:440-5. http://www.ncbi.nlm.nih.gov/pubmed/10335703?dopt=AbstractPlus

555. Mendelman PM, Del Beccaro MA, McLinn SE et al. Cefpodoxime proxetil compared with amoxicillin-clavulanate for the treatment of otitis media. J Pediatr. 1992; 121:459-65. http://www.ncbi.nlm.nih.gov/pubmed/1517926?dopt=AbstractPlus

556. Arguedas AG, Zaleska M, Stutman HR et al. Comparative trial of cefprozil vs. amoxicillin clavulanate potassium in the treatment of children with acute otitis media with effusion. Pediatr Infect Dis J. 1991; 10:375-80. http://www.ncbi.nlm.nih.gov/pubmed/1906160?dopt=AbstractPlus

557. Giebink GS, Canafax DM, Kempthorne J. Antimicrobial treatment of acute otitis media. J Pediatr. 1991; 119:495-500. http://www.ncbi.nlm.nih.gov/pubmed/1880671?dopt=AbstractPlus

558. Stool SE, Berg AO, Berman S et al for the Otitis Media Guideline Panel. Otitis media with effusion in young children. Clinical Practice Guideline. Number 12. AHCPR Publication No. 94-0622. Rockville, MD: Agency for Health Care Policy and Research, Public Health Service, US Department of Health and Human Resources. July 1994.

559. Berman S. Otitis media in children. N Engl J Med. 1995; 332:1560-5. http://www.ncbi.nlm.nih.gov/pubmed/7739711?dopt=AbstractPlus

560. Bluestone CD. Current therapy for otitis media and criteria for evaluation of new antimicrobial agents. Clin Infect Dis. 1992; 14(Suppl 2):S197-203. http://www.ncbi.nlm.nih.gov/pubmed/1617038?dopt=AbstractPlus

561. White LL, Holimon TD, Tepedino JT et al. Antimicrobials prescribed for otitis media in a pediatric Medicaid population. Am J Health-Syst Pharm. 1996; 53:2963-9. http://www.ncbi.nlm.nih.gov/pubmed/8974159?dopt=AbstractPlus

562. Poole MD. Otitis media complications and treatment failures: implications of pneumococcal resistance. Pediatr Infect Dis J. 1995; 14(suppl):S23-9. http://www.ncbi.nlm.nih.gov/pubmed/7792127?dopt=AbstractPlus

563. McLinn S, Williams D. Incidence of antibiotic-resistant Streptococcus pneumoniae and beta-lactamase-positive Haemophilus influenzae. in clinical isolates from patients with otitis media. Pediatr Infect Dis J. 1996; 15(Suppl):S3-9. http://www.ncbi.nlm.nih.gov/pubmed/8878239?dopt=AbstractPlus

564. Patel JA, Reisner B, Vizirinia N et al. Bacteriologic failure of amoxicillin-clavulanate in treatment of acute otitis media caused by nontypeable Haemophilus influenzae . J Pediatr. 1995; 126:799-806. http://www.ncbi.nlm.nih.gov/pubmed/7752010?dopt=AbstractPlus

565. Pichichero ME. Assessing the treatment alternatives for acute otitis media. Pediatr Infect Dis J. 1994; 13(Suppl):S27-34.

566. McLinn SE, McCarty JM, Perrotta R et al. Multicenter controlled trial comparing ceftibuten with amoxicillin/clavulanate in the empiric treatment of acute otitis media. Pediatr Infect Dis J. 1995; 14(Suppl):S108-14.

567. Berman S, Roark R. Factors influencing outcome in children treated with antibiotics for acute otitis media. Pediatr Infect Dis J. 1993; 12:20-4. http://www.ncbi.nlm.nih.gov/pubmed/8417420?dopt=AbstractPlus

568. Block SL. Causative pathogens, antibiotic resistance and therapeutic considerations in acute otitis media. Pediatr Infect Dis J. 1997; 16:449-56. http://www.ncbi.nlm.nih.gov/pubmed/9109158?dopt=AbstractPlus

569. Block SL, Harrison CJ, Hedrick JA et al. Penicillin-resistant Streptococcus pneumoniae in acute otitis media: risk factors, susceptibility patterns and antimicrobial management. Pediatr Infect Dis J. 1995; 14:751-9. http://www.ncbi.nlm.nih.gov/pubmed/8559623?dopt=AbstractPlus

570. Guerrant RL, Gilder TV, Steiner TS et al. Practice guidelines for the management of infectious diarrhea. Clin Infect Dis. 2001; 32:331-50. http://www.ncbi.nlm.nih.gov/pubmed/11170940?dopt=AbstractPlus

571. Boccazzi A, Careddu P. Acute otitis media in pediatrics: are there rational issues for empiric therapy? Pediatr Infect Dis J. 1997; 16(Suppl):S65-9.

572. Paradise JL. Treatment guidelines for otitis media: the need for breadth and flexibility. Pediatr Infect Dis J. 1995; 14:429-35. http://www.ncbi.nlm.nih.gov/pubmed/7638033?dopt=AbstractPlus

573. Mandel EM, Casselbrant ML, Kurs-Lasky M et al. Efficacy of ceftibuten compared with amoxicillin for otitis media with effusion in infants and children. Pediatr Infect Dis J. 1996; 15:409-14. http://www.ncbi.nlm.nih.gov/pubmed/8724062?dopt=AbstractPlus

574. Oh PI, Maerov P, Pritchard D et al. A cost-utility analysis of second-line antibiotics in the treatment of acute otitis media in children. Clin Ther. 1996; 18:160-82. http://www.ncbi.nlm.nih.gov/pubmed/8851461?dopt=AbstractPlus

575. Berman S, Roark R, Luckey D. Theoretical cost effectiveness of management options for children with persisting middle ear effusions. Pediatrics. 1994; 93:353-63. http://www.ncbi.nlm.nih.gov/pubmed/8115191?dopt=AbstractPlus

576. McLinn SE, Moskal M, Goldfarb J et al. Comparison of cefuroxime axetil and amoxicillin-clavulanate suspensions in treatment of acute otitis media with effusion in children. Antimicrob Agents Chemother. 1994; 38:315-8. http://www.ncbi.nlm.nih.gov/pubmed/8192458?dopt=AbstractPlus

577. Kaplan B, Wandstrat TL, Cunningham JR. Overall cost in the treatment of otitis media. Pediatr Infect Dis J. 1997; 16(Suppl):S9-11. http://www.ncbi.nlm.nih.gov/pubmed/9041621?dopt=AbstractPlus

578. Mandel EM, Casselbrandt ML, Rockette HE et al. Efficacy of 20- versus 10-day antimicrobial treatment for acute otitis media. Pediatrics. 1995; 96:5-13. http://www.ncbi.nlm.nih.gov/pubmed/7596722?dopt=AbstractPlus

579. Marchisio P, Principi N, Sorella S et al. Etiology of acute otitis media in human immunodeficiency virus-infected children. Pediatr Infect Dis J. 1996; 15:58-61. http://www.ncbi.nlm.nih.gov/pubmed/8684878?dopt=AbstractPlus

580. Shulman ST, Bisno AL, Clegg HW et al. Clinical practice guideline for the diagnosis and management of group A streptococcal pharyngitis: 2012 update by the Infectious Diseases Society of America. Clin Infect Dis. 2012; 55:1279-82. http://www.ncbi.nlm.nih.gov/pubmed/23091044?dopt=AbstractPlus

581. Gooch WM, Blair E, Puopolo A et al. Effectiveness of five days of therapy with cefuroxime axetil suspension for the treatment of acute otitis media. Pediatr Infect Dis J. 1996; 15:157-64. http://www.ncbi.nlm.nih.gov/pubmed/8822290?dopt=AbstractPlus

582. Anon. Vibrio vulnificus infections associated with eating raw oysters—Los Angeles, 1996. MMWR Morb Mortal Wkly Rep. 1996; 45:621-4. http://www.ncbi.nlm.nih.gov/pubmed/8965788?dopt=AbstractPlus

583. Del Castillo F, Baquero-Artigao F, Garcia-Perea A. Influence of recent antibiotic therapy on antimicrobial resistance of Streptococcus pneumoniae in children with acute otitis media in Spain. Pediatr Infect Dis J. 1998; 17:94-7. http://www.ncbi.nlm.nih.gov/pubmed/9493802?dopt=AbstractPlus

584. Dagan R, Abramson O, Leibovitz E et al. Impaired bacteriologic response to oral cephalosporins in acute otitis media caused by pneumococci with intermediate resistance to penicillin. Pediatr Infect Dis J. 1996; 15:980-5. http://www.ncbi.nlm.nih.gov/pubmed/8933545?dopt=AbstractPlus

585. Dagan R, Abramson O, Leibovitz E et al. Bacteriologic response to oral cephalosporins: are established susceptibility breakpoints appropriate in the case of acute otitis media? J Infect Dis. 1997; 176:1253-9.

586. Pitkaranta A, Virolainen A, Jero J et al. Detection of rhinovirus, respiratory syncytial virus, and coronavirus infections in acute otitis media by reverse transcriptase polymerase chain reaction. Pediatrics. 1998; 102:291-5. http://www.ncbi.nlm.nih.gov/pubmed/9685428?dopt=AbstractPlus

587. Arola M, Ziegler T, Ruuskanen O. Respiratory virus infection as a cause of prolonged symptoms in acute otitis media. J Pediatr. 1990; 116:697-701. http://www.ncbi.nlm.nih.gov/pubmed/2329417?dopt=AbstractPlus

588. Culpepper L, Froom J. Routine antimicrobial treatment of acute otitis media: is it necessary? JAMA. 1997; 278:1643-5.

590. Cantekin EI. The changing treatment paradigm for acute otitis media. JAMA. 1998; 280:1903. http://www.ncbi.nlm.nih.gov/pubmed/9851459?dopt=AbstractPlus

591. Moffatt MEK, Koxyrskyi AL, Klassen TP et al. The changing treatment paradigm of acute otitis media. JAMA. 1998; 280:1903-4.

592. Fahey T, Sharp D. Acute otitis media in children: reappraisal of management of acute otitis media is required. BMJ. 1998; 316:630. http://www.ncbi.nlm.nih.gov/pubmed/9518937?dopt=AbstractPlus

593. Majeed A, Harris T. Acute otitis media in children. BMJ. 1997; 315:321-2. http://www.ncbi.nlm.nih.gov/pubmed/9270442?dopt=AbstractPlus

594. Froom J, Culpepper L, Jacobs M et al. Antimicrobials for acute otitis media? A review from the International Primary Care Network. BMJ. 1997; 315:98-102. http://www.ncbi.nlm.nih.gov/pubmed/9240050?dopt=AbstractPlus

595. Del Mar C, Glasziou P, Hayem M. Are antibiotics indicated as initial treatment for children with acute otitis media? A meta-analysis. BMJ. 1997; 314:1526-9. http://www.ncbi.nlm.nih.gov/pubmed/9183201?dopt=AbstractPlus

596. Arason VA, Kristinsson KG, Sigurdsson JA et al. Do antimicrobials increase the carriage rate of penicillin resistant pneumococci in children? Cross sectional prevalence study. BMJ. 1996; 313:387-91. http://www.ncbi.nlm.nih.gov/pubmed/8761224?dopt=AbstractPlus

597. Maw R, Bawden R. Spontaneous resolution of severe chronic glue ear in children and the effect of adenoidectomy, tonsillectomy, and insertion of ventilation tubes (grommets). BMJ. 1993; 306:756-60. http://www.ncbi.nlm.nih.gov/pubmed/8490338?dopt=AbstractPlus

598. Arguedas A, Loaiza C, Herrera JF et al. Antimicrobial therapy for children with chronic suppurative otitis media without cholesteatoma. Pediatr Infect Dis J. 1994; 13:878-82. http://www.ncbi.nlm.nih.gov/pubmed/7854886?dopt=AbstractPlus

599. Brook I. Management of chronic suppurative otitis media: superiority of therapy effective against anaerobic bacteria. Pediatr Infect Dis J. 1994; 13:188-93. http://www.ncbi.nlm.nih.gov/pubmed/8177625?dopt=AbstractPlus

600. Arguedas AG, Herrera JF, Faingezicht I et al. Ceftazidime for therapy of children with chronic suppurative otitis media without cholesteatoma. Pediatr Infect Dis J. 1993; 12:246-8. http://www.ncbi.nlm.nih.gov/pubmed/8451104?dopt=AbstractPlus

601. Dagan R, Fliss DM, Einihorn M et al. Outpatient management of chronic suppurative otitis media without cholesteatoma in children. Pediatr Infect Dis J. 1992; 11:542-6. http://www.ncbi.nlm.nih.gov/pubmed/1528644?dopt=AbstractPlus

602. Eppes SC, Klein DJ, Lewis LL. Ceftriaxone for acute otitis media. Pediatrics. 1997; 100:157. http://www.ncbi.nlm.nih.gov/pubmed/9229709?dopt=AbstractPlus

603. Cantekin EJ. Ceftriaxone for acute otitis media. Pediatrics. 1997; 100:157-8.

604. Barnett ED, Teele DW, Klein JO et al. Ceftriaxone for acute otitis media. Pediatrics. 1997; 100:158. http://www.ncbi.nlm.nih.gov/pubmed/9229710?dopt=AbstractPlus

605. Johnson MP, Ramphal R. Malignant external otitis: report on therapy with ceftazidime and review of therapy and prognosis. Clin Infect Dis. 1990; 12:173-80.

606. Cates C. An evidence based approach to reducing antibiotic use in children with acute otitis media: controlled before and after study. BMJ. 1999; 318:715-6. http://www.ncbi.nlm.nih.gov/pubmed/10074019?dopt=AbstractPlus

607. Preston D, Therasse D, Zeckel ML. Cefaclor and cefuroxime axetil in the treatment of otitis media—an alternative view. J Infect Dis. 1998; 178:1547-8. http://www.ncbi.nlm.nih.gov/pubmed/9780287?dopt=AbstractPlus

608. Bluestone CD. Epidemiology and pathogenesis of chronic suppurative otitis media: implications for prevention and treatment. Int J Pediatr Otorhinolaryngology. 1998; 42:207-23.

609. Kafetzis DA. Multi-investigator evaluation of the efficacy and safety of cefprozil, amoxicillin-clavulanate, cefixime and cefaclor in the treatment of acute otitis media, Eur J Clin Microbiol Infect Dis. 1994; 13:857-65.

610. Adler M, McDonald PJ, Trostmann U et al. Cefdinir versus amoxicillin/clavulanic acid in the treatment of suppurative acute otitis media in children. Eur J Clin Microbiol Infect Dis. 1997; 16:214-9. http://www.ncbi.nlm.nih.gov/pubmed/9131324?dopt=AbstractPlus

611. Hsu GS, Levine SC, Giebink GS. Management of otitis media using agency for health care policy and research guidelines. Otolaryngol Head Neck Surg. 1998; 118:437-43. http://www.ncbi.nlm.nih.gov/pubmed/9560092?dopt=AbstractPlus

612. Perry BP, Zieno SA, Yonkers AJ. Outcome-oriented managed care comparing efficacies of cefaclor and amoxicillin in acute and recurrent acute otitis media. Ear Nose Throat J. 1995; 74:840-4. http://www.ncbi.nlm.nih.gov/pubmed/8556984?dopt=AbstractPlus

613. Leibovitz E, Piglansky l, Raiz S et al. Bacteriologic efficacy of a three-day intramuscular ceftriaxone regimen in nonresponsive acute otitis media. Pediatr Infect Dis J. 1998; 17:1126-31. http://www.ncbi.nlm.nih.gov/pubmed/9877360?dopt=AbstractPlus

614. 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; 18:403-9. http://www.ncbi.nlm.nih.gov/pubmed/10353511?dopt=AbstractPlus

615. Ball P. Therapy for pneumococcal infection at the millennium: doubts and certainties. Am J Med. 1999; 107(Suppl 1A):77S-85S. http://www.ncbi.nlm.nih.gov/pubmed/10451013?dopt=AbstractPlus

616. Gooch WM, Adelglass J, Kelsey DK et al. Loracarbef versus clarithromycin in children with acute otitis media with effusion. Clin Ther. 1999; 21:711-21. http://www.ncbi.nlm.nih.gov/pubmed/10363736?dopt=AbstractPlus

617. Adderson EE. Preventing otitis media: medial approaches. Pediatr Ann. 1998; 27:101-7. http://www.ncbi.nlm.nih.gov/pubmed/9523298?dopt=AbstractPlus

618. Bluestone CD, Klein JO. Chronic suppurative otitis media. Pediatr Rev. 1999; 20:277-9. http://www.ncbi.nlm.nih.gov/pubmed/10429148?dopt=AbstractPlus

619. Hoppe HL, Johnson CE. Otitis media: focus on antimicrobial resistance and new treatment options. Am J Health-Syst Pharm. 1998; 55:1881-97. http://www.ncbi.nlm.nih.gov/pubmed/9784768?dopt=AbstractPlus

620. Maw R, Wilks J, Harvey I et al. Early surgery compared with watchful waiting for glue ear and effect on language development in preschool children: a randomised trial. Lancet. 1999; 353:960-3. http://www.ncbi.nlm.nih.gov/pubmed/10459904?dopt=AbstractPlus

621. Subba Rao SD, Macias MP, Dillman CA et al. A randomized, observer-blind trial of amoxycillin/clavulanate versus cefaclor in the treatment of children with acute otitis media. J Chemother. 1998; 10:460-8. http://www.ncbi.nlm.nih.gov/pubmed/9876054?dopt=AbstractPlus

622. Bluestone CD. Ear and mastoid infections. In: Gorbach SL, Bartlett JG, Blacklow NR, eds. Infectious Diseases. Philadelphia, PA: WB Saunders; 1998:530-9.

623. Sawaya GF, Grady D, Kerlikowske K et al. Antibiotics at the time of induced abortion: the case for universal prophylaxis based on a meta-analysis. Obstet Gynecol. 1996; 87:884-90. http://www.ncbi.nlm.nih.gov/pubmed/8677129?dopt=AbstractPlus

625. Shammo JM, Calhoun B, Mauer AM et al. First two cases of immune hemolytic anemia associated with ceftizoxime. Transfusion. 1999; 39:838-44. http://www.ncbi.nlm.nih.gov/pubmed/10504119?dopt=AbstractPlus

626. Moallem HJ, Garratty G, Wakeham M et al. Ceftriaxone-related fatal hemolysis in an adolescent with perinatally acquired human immunodeficiency virus infection. J Pediatr. 1998; 133:279-81. http://www.ncbi.nlm.nih.gov/pubmed/9709722?dopt=AbstractPlus

627. Shulman IA, Arndt PA, McGehee W et al. Cefotaxime-induced immune hemolytic anemia due to antibodies reacting in vitro by more than one mechanism. Transfusion. 1990; 30:263-6. http://www.ncbi.nlm.nih.gov/pubmed/2316002?dopt=AbstractPlus

628. Garratty G, Leger RM, Arndt PA. Severe immune hemolytic anemia associated with prophylactic use of cefotetan in obstetric and gynecologic procedures. Am J Obstet Gynecol. 1999; 181:103-4. http://www.ncbi.nlm.nih.gov/pubmed/10411803?dopt=AbstractPlus

629. Noyes N, Berkeley AS, Freedman K et al. Incidence of postpartum endomyometritis following single-dose antibiotic prophylaxis with either ampicillin/sulbactam, cefazolin, or cefotetan in high-risk cesarean section patients. Infect Dis Obstet Gynecol. 1998; 6:220-3. http://www.ncbi.nlm.nih.gov/pubmed/9894177?dopt=AbstractPlus

630. Goldblatt EL, Dohar J, Nozza RJ et al. Topical ofloxacin versus systemic amoxicillin/clavulanate in purulent otorrhea in children with tympanostomy tubes. Int J Pediatr Otorhinolaryngology. 1998:91-101.

631. Helpern MT, Palmer CS, Seidlin M. Treatment patterns for otitis externa. J Am Board Fam Pract. 1999; 12:1-7. http://www.ncbi.nlm.nih.gov/pubmed/10050637?dopt=AbstractPlus

632. Hern JD, Ghufoor K, Jayaraj SM et al. ENT manifestations of Pseudomonas aeruginosa infection in HIV and AIDS. Int J Clin Pract. 1998; 52:141-4. http://www.ncbi.nlm.nih.gov/pubmed/9684426?dopt=AbstractPlus

633. Pessey JJ, Gehanno P, Thoroddsen E et al. Short course therapy with cefuroxime axetil for acute otitis media: results of a randomized multicenter comparison with amoxicillin/clavulanate. Pediatr Infect Dis J. 1999; 18:854-9. http://www.ncbi.nlm.nih.gov/pubmed/10530579?dopt=AbstractPlus

634. Beswick AJ, Lawley B, Fraise AP et al. Detection of Alliococcus otitis in mixed bacterial populations from middle-ear effusions of patients with otitis media. Lancet. 1999; 354:386-9. http://www.ncbi.nlm.nih.gov/pubmed/10437868?dopt=AbstractPlus

635. Higgins A, London J, Charland S et al. Prophylactic antibiotics for elective laparoscopic cholecystectomy: are they necessary? Arch Surg. 1999; 134:611-3.

636. Mandel EM, Rockette HE, Paradise JL et al. Comparative efficacy of erythromycin-sulfisoxazole, cefaclor, amoxicillin or placebo for otitis media with effusion in children. Pediatr Infect Dis J. 1991; 10:899-906. http://www.ncbi.nlm.nih.gov/pubmed/1766705?dopt=AbstractPlus

637. Nichols Rl, Smith JW, Garcia RY et al. Current practices of preoperative bowel preparation among North American colorectal surgeons. Clin Infect Dis. 1997; 24:609-19. http://www.ncbi.nlm.nih.gov/pubmed/9145734?dopt=AbstractPlus

638. Schoetz DJ, Roberts PL, Murray JJ et al. Addition of parenteral cefoxitin to regimen of oral antibiotics for elective colorectal operations. A randomized prospective study. Ann Surg. 1990; 212:209-12. http://www.ncbi.nlm.nih.gov/pubmed/2100983?dopt=AbstractPlus

639. Da Costa A, Kirkorian G, Cucherat M et al. Antibiotic prophylaxis for permanent pacemaker implantation: a meta-analysis. Circulation. 1998; 97:1796-801. http://www.ncbi.nlm.nih.gov/pubmed/9603534?dopt=AbstractPlus

640. Gonzalez RP, Holevar MR. Role of prophylactic antibiotics for tube thoracostomy in chest trauma. Am Surg. 1998; 64:617-20. http://www.ncbi.nlm.nih.gov/pubmed/9655270?dopt=AbstractPlus

641. Aznar R, Mateu M, Miro JM et al. Antibiotic prophylaxis in non-cardiac thoracic surgery: cefazolin versus placebo. Eur J Cardiothorac Surg. 19991; 5:515-8.

642. Scher KS. Studies on the duration of antibiotic administration for surgical prophylaxis. Am Surg. 1997; 63:59-62. http://www.ncbi.nlm.nih.gov/pubmed/8985073?dopt=AbstractPlus

643. Nooyen SM, Overbeek BP, de la Riviere B et al. Prospective randomised comparison of single-dose versus multiple-dose cefuroxime for prophylaxis in coronary artery bypass grafting. Eur J Clin Microbiol Infect Dis. 1994; 13:1033-7. http://www.ncbi.nlm.nih.gov/pubmed/7889965?dopt=AbstractPlus

644. Hall JC, Christiansen K, Carter MJ et al. Antibiotic prophylaxis in cardiac surgery. Ann Thorac Surg. 1993; 56:916-22. http://www.ncbi.nlm.nih.gov/pubmed/8215669?dopt=AbstractPlus

645. Roark R, Berman S. Continuous twice daily or once daily amoxicillin prophylaxis compared with placebo for children with recurrent acute otitis media. Pediatr Infect Dis J. 1997; 16:376-81. http://www.ncbi.nlm.nih.gov/pubmed/9109139?dopt=AbstractPlus

646. Hendolin PH, Karkkainen U, Himi T et al. High incidence of Alloiococcus otitis in otitis media with effusion. Pediatr Infect Dis J. 1999; 18:860-5. http://www.ncbi.nlm.nih.gov/pubmed/10530580?dopt=AbstractPlus

649. Nowakowski J, Schwartz I, Nadelman RB et al. Culture-confirmed infection and reinfection with Borrelia burgdorferi . Ann Intern Med. 1997; 127:130-2. http://www.ncbi.nlm.nih.gov/pubmed/9230002?dopt=AbstractPlus

650. Steere AC. Borrelia burgdorferi (Lyme Disease, Lyme Borreliosis). In: Mandell GL, Bennett JE, Dolin R eds. Mandell, Douglas and Bennett’s principles and practices of infectious disease. 4th ed. New York: Churchill Livingstone; 1995:2143-55.

651. Shadick NA, Phillips CB, Logigian EL et al. The long-term clinical outcomes of Lyme disease: a population-based retrospective cohort study. Ann Intern Med. 1994; 121:560-7. http://www.ncbi.nlm.nih.gov/pubmed/8085687?dopt=AbstractPlus

652. Tugwell P, Dennis DT, Weinstein A et al. Laboratory evaluation in the diagnosis of Lyme disease. Ann Intern Med. 1997; 127:1109-23. http://www.ncbi.nlm.nih.gov/pubmed/9412316?dopt=AbstractPlus

653. Dressler F, Whalen JA, Reinhard BN et al. Western blotting in the serodiagnosis of Lyme disease. J Infect Dis. 1993; 167:392-400. http://www.ncbi.nlm.nih.gov/pubmed/8380611?dopt=AbstractPlus

654. Johnson BJB, Robbins KE, Bailey RE et al. Serodiagnosis of Lyme disease: accuracy of a two-step approach using a flagella-based ELISA and immunoblotting. J Infect Dis. 1996; 174:346-53. http://www.ncbi.nlm.nih.gov/pubmed/8699065?dopt=AbstractPlus

655. Fikrig E, Huguenel ED, Berland R et al. Serologic diagnosis of Lyme disease using recombinant outer surface proteins A and B and flagellin. J Infect Dis. 1992; 165:1127-32. http://www.ncbi.nlm.nih.gov/pubmed/1583333?dopt=AbstractPlus

656. Fix AD, Strickland GT, Grant J. Tick bites and Lyme disease in an endemic setting: problematic use of serologic testing and prophylactic antibiotic therapy. JAMA. 1998; 279:206-10. http://www.ncbi.nlm.nih.gov/pubmed/9438740?dopt=AbstractPlus

657. Barbour AG. Expert advice and patient expectations: laboratory testing and antibiotics for Lyme disease. JAMA. 1998; 279:239-40. http://www.ncbi.nlm.nih.gov/pubmed/9438748?dopt=AbstractPlus

658. Hilton E, DeVoti J, Benach JL et al. Seroprevalence and seroconversion for tick-borne diseases in a high-risk population in the northeast United States. Am J Med. 1999; 106:404-9. http://www.ncbi.nlm.nih.gov/pubmed/10225242?dopt=AbstractPlus

659. Trevejo RT, Krause PJ, Sikand VK et al. Evaluation of two-test serodiagnostic method for early Lyme disease in clinical practice. J Infect Dis. 1999; 179:931-8. http://www.ncbi.nlm.nih.gov/pubmed/10068589?dopt=AbstractPlus

660. Porwancher R. A reanalysis of IgM western blot criteria for the diagnosis of early Lyme disease. J Infect Dis. 1999; 179:1021-4. http://www.ncbi.nlm.nih.gov/pubmed/10068602?dopt=AbstractPlus

661. Dennis DT. Lyme disease. Dermatol Clin. 1995; 13:537-51. http://www.ncbi.nlm.nih.gov/pubmed/7554502?dopt=AbstractPlus

663. Vansen Pharma Inc. Spectracef (cefditoren pivoxil) tablets prescribing information. Westmount, QC; 2011.

664. Kuti JL, Quintiliani R. Cefditoren pivoxil: a novel broad-spectrum oral cephalosporin. Formulary. 2001; 36:265-75.

666. Steere AC. Lyme disease. N Engl J Med. 2001; 345:115-25. http://www.ncbi.nlm.nih.gov/pubmed/11450660?dopt=AbstractPlus

667. Patel R, Grogg KL, Edwards WD et al. Death from inappropriate therapy for Lyme disease. Clin Infect Dis. 2000; 31:1107-9. http://www.ncbi.nlm.nih.gov/pubmed/11049799?dopt=AbstractPlus

668. Klempner MS, Hu LT, Evans J et al. Two controlled trials of antibiotic treatment in patients with persistent symptoms and a history of Lyme disease. N Engl J Med. 2001; 345:85-92. http://www.ncbi.nlm.nih.gov/pubmed/11450676?dopt=AbstractPlus

669. Thompson C, Spielman A, Krause P. Coinfecting deer-associated zoonoses: Lyme disease, babesiosis, and ehrlichiosis. Clin Infect Dis. 2000; 33:676-85.

670. Sigal LH. Lyme disease: primum non nocere. J Infect Dis. 1995; 171:423-4. http://www.ncbi.nlm.nih.gov/pubmed/7844380?dopt=AbstractPlus

671. Nowakowski J, McKenna D, Nadelman RB et al. Failure of treatment with cephalexin for Lyme disease. Arch Fam Med. 2000; 9:563-7. http://www.ncbi.nlm.nih.gov/pubmed/10862221?dopt=AbstractPlus

672. Dotevall L, Alestig K, Hanner P et al. The use of doxycycline in nervous system Borrelia burgdorferi infection. Scand J Infect Dis Suppl. 1988; 53:74-9. http://www.ncbi.nlm.nih.gov/pubmed/3166545?dopt=AbstractPlus

673. Eidlitz-Marcus T, Cohen YH, Nussinovitch M et al. Comparative efficacy of two- and five-day courses of ceftriaxone for treatment of severe shigellosis in children. J Pediatr. 1993; 123:822-4. http://www.ncbi.nlm.nih.gov/pubmed/8229499?dopt=AbstractPlus

674. Varsano I, Eidlitz-Marcus T, Nussinovitch M et al. Comparative efficacy of ceftriaxone and ampicillin for treatment of severe shigellosis in children. J Pediatr. 1991; 188:627-32.

675. Abdel-Haq NM, Asmar BI, Abuhammour WM et al. Yersinia enterocolitica infection in children. Pediatr Infect Dis J. 2000; 19:954-8. http://www.ncbi.nlm.nih.gov/pubmed/11055595?dopt=AbstractPlus

676. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing: Twenty-first informational supplement. CLSI document M100-S21. Wayne, PA; 2011.

677. Lupin Pharma. Suprax (cefixime) tablets, capsules, chewable tablets, and powder for oral suspension prescribing information. Baltimore, MD; 2013 Mar.

678. Cooper RJ, Hoffman JR, Bartlett JG et al. Principles of appropriate antibiotic use for acute pharyngitis in adults: background. Ann Intern Med. 2001; 134:509-17. http://www.ncbi.nlm.nih.gov/pubmed/11255530?dopt=AbstractPlus

680. Mandell LA, Wunderink RG, Anzueto A et al. 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. http://www.ncbi.nlm.nih.gov/pubmed/17278083?dopt=AbstractPlus

682. Heffelfinger, JD, Dowell SF 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; 160:1399-1408. http://www.ncbi.nlm.nih.gov/pubmed/10826451?dopt=AbstractPlus

683. Lieberthal AS, Carroll AE, Chonmaitree T et al. The diagnosis and management of acute otitis media. Pediatrics. 2013; 131:e964-99. http://www.ncbi.nlm.nih.gov/pubmed/23439909?dopt=AbstractPlus

684. American Academy of Pediatrics, American Academy of Family Physicians, American Academy of Otolaryngology-Head and Neck Surgery, and American Academy of Pediatrics Subcommittee on Otitis Media with Effusion. Otitis media with effusion. Pediatrics. 2004: 113:1412-29.

685. Ferrieri P, Gewitz MH, Gerber MA et al. Unique features of infective endocarditis in childhood. Circulation. 2002; 105:2115-27. http://www.ncbi.nlm.nih.gov/pubmed/11980694?dopt=AbstractPlus

686. Pfister HW, Preac-Mursic V, Wilske B et al. Randomized comparison of ceftriaxone and cefotaxime in Lyme neuroborreliosis. J Infect Dis. 1991; 163:311-8. http://www.ncbi.nlm.nih.gov/pubmed/1988514?dopt=AbstractPlus

687. Centers for Disease Control and Prevention. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever, ehrlichioses, and anaplasmosis -United States: a practical guide for physicians and other health-care and public health professionals.. MMWR Recomm Rep. 2006; 55(RR-4): 1-27. http://www.cdc.gov/mmwr/PDF/rr/rr5504.pdf

690. Panaphut T, Domrongkitchaiporn S, Vibhagool A et al. Ceftriaxone compared with sodium penicillin g for treatment of severe leptospirosis. Clin Infect Dis. 2003; 36:1507-13. http://www.ncbi.nlm.nih.gov/pubmed/12802748?dopt=AbstractPlus

691. Farr RW. Leptospirosis. Clin Infect Dis. 1995; 21:1-6; quiz 7-8. http://www.ncbi.nlm.nih.gov/pubmed/7578715?dopt=AbstractPlus

693. Suputtamongkol Y, Niwattayakul K, Suttinont C et al. An open, randomized, controlled trial of penicillin, doxycycline, and cefotaxime for patients with severe leptospirosis. Clin Infect Dis. 2004; 39:1417-24. http://www.ncbi.nlm.nih.gov/pubmed/15546074?dopt=AbstractPlus

694. Katz AR, Ansdell VE, Effler PV et al. Assessment of the clinical presentation and treatment of 353 cases of laboratory-confirmed leptospirosis in Hawaii, 1974-1998. Clin Infect Dis. 2001; 33:1834-41. http://www.ncbi.nlm.nih.gov/pubmed/11692294?dopt=AbstractPlus

695. Griffith ME, Hospenthal DR, Murray CK. Antimicrobial therapy of leptospirosis. Curr Opin Infect Dis. 2006; 19:533-7. http://www.ncbi.nlm.nih.gov/pubmed/17075327?dopt=AbstractPlus

696. Ressner RA, Griffith ME, Beckius ML et al. Antimicrobial susceptibilities of geographically diverse clinical human isolates of Leptospira. Antimicrob Agents Chemother. 2008; 52:2750-4. http://www.ncbi.nlm.nih.gov/pubmed/18411316?dopt=AbstractPlus

697. van Burik JA, Hackman RC, Nadeem SQ et al. Nocardiosis after bone marrow transplantation: a retrospective study. Clin Infect Dis. 1997; 24:1154-60. http://www.ncbi.nlm.nih.gov/pubmed/9195074?dopt=AbstractPlus

698. Garlando F, Bodmer T, Lee C et al. Successful treatment of disseminated nocardiosis complicated by cerebral abscess with ceftriaxone and amikacin: case report. Clin Infect Dis. 1992; 15:1039-40. http://www.ncbi.nlm.nih.gov/pubmed/1457635?dopt=AbstractPlus

699. Garcia del Palacio JI, Martín Pérez I. Response of pulmonary nocardiosis to ceftriaxone in a patient with AIDS. Chest. 1993; 103:1925-6. http://www.ncbi.nlm.nih.gov/pubmed/8404142?dopt=AbstractPlus

700. Unkle DW, Ricketti AJ, Cleri DJ et al. An HIV-infected patient with Nocardia asteroides bilateral pneumonia. AIDS Read. 2008; 18:566-8. http://www.ncbi.nlm.nih.gov/pubmed/19062401?dopt=AbstractPlus

701. McPhee L, Stogsdill P, Vella JP. Nocardia farcinica pericarditis after kidney transplantation despite prophylaxis. Transpl Infect Dis. 2009; :. http://www.ncbi.nlm.nih.gov/pubmed/19508699?dopt=AbstractPlus

702. Kilincer C, Hamamcioglu MK, Simsek O et al. Nocardial brain abscess: review of clinical management. J Clin Neurosci. 2006; 13:481-5. http://www.ncbi.nlm.nih.gov/pubmed/16678731?dopt=AbstractPlus

703. Forest Pharmaceuticals, Inc. Teflaro (ceftaroline fosamil) for injection prescribing information. St. Louis, MO; 2011 Jan.

704. Zhanel GG, Sniezek G, Schweizer F et al. Ceftaroline: a novel broad-spectrum cephalosporin with activity against meticillin-resistant Staphylococcus aureus. Drugs. 2009; 69:809-31. http://www.ncbi.nlm.nih.gov/pubmed/19441869?dopt=AbstractPlus

705. Kollef MH. New antimicrobial agents for methicillin-resistant Staphylococcus aureus. Crit Care Resusc. 2009; 11:282-6. http://www.ncbi.nlm.nih.gov/pubmed/20001879?dopt=AbstractPlus

706. Schirmer PL, Deresinski SC. Ceftobiprole: a new cephalosporin for the treatment of skin and skin structure infections. Expert Rev Anti Infect Ther. 2009; 7:777-91. http://www.ncbi.nlm.nih.gov/pubmed/19735220?dopt=AbstractPlus

708. Solomkin JS, Mazuski JE, Bradley JS et al. Diagnosis and management of complicated intra-abdominal infection in adults and children: guidelines by the Surgical Infection Society and the Infectious Diseases Society of America. Clin Infect Dis. 2010; 50:133-64. http://www.ncbi.nlm.nih.gov/pubmed/20034345?dopt=AbstractPlus

710. . Drugs for bacterial infections. Treat Guidel Med Lett. 2010; 8:43-52. http://www.ncbi.nlm.nih.gov/pubmed/20489679?dopt=AbstractPlus

711. Skrupky LP, Micek ST, Kollef MH. Bench-to-bedside review: Understanding the impact of resistance and virulence factors on methicillin-resistant Staphylococcus aureus infections in the intensive care unit. Crit Care. 2009; 13:222. http://www.ncbi.nlm.nih.gov/pubmed/19889197?dopt=AbstractPlus

712. Ritchie DJ, Alexander BT, Finnegan PM. New antimicrobial agents for use in the intensive care unit. Infect Dis Clin North Am. 2009; 23:665-81. http://www.ncbi.nlm.nih.gov/pubmed/19665089?dopt=AbstractPlus

713. Saravolatz L, Pawlak J, Johnson L. In vitro activity of ceftaroline against community-associated methicillin-resistant, vancomycin-intermediate, vancomycin-resistant, and daptomycin-nonsusceptible Staphylococcus aureus isolates. Antimicrob Agents Chemother. 2010; 54:3027-30. http://www.ncbi.nlm.nih.gov/pubmed/20404122?dopt=AbstractPlus

714. Ge Y, Biek D, Talbot GH et al. In vitro profiling of ceftaroline against a collection of recent bacterial clinical isolates from across the United States. Antimicrob Agents Chemother. 2008; 52:3398-407. http://www.ncbi.nlm.nih.gov/pubmed/18625769?dopt=AbstractPlus

715. Andes DR, Craig WA. Cephalosporins. In: Bennett JE, Dolin R, Blaser MJ eds. Mandell, Douglas and Bennett’s principles and practices of infectious disease. 8th ed. Philadelphia, PA: Saunders, Elsevier; 2015:278-91.

716. Centers for Disease Control and Prevention (CDC). Cephalosporin susceptibility among Neisseria gonorrhoeae isolates--United States, 2000-2010. MMWR Morb Mortal Wkly Rep. 2011; 60:873-7. http://www.ncbi.nlm.nih.gov/pubmed/21734634?dopt=AbstractPlus

717. Ohnishi M, Golparian D, Shimuta K et al. Is Neisseria gonorrhoeae initiating a future era of untreatable gonorrhea?: detailed characterization of the first strain with high-level resistance to ceftriaxone. Antimicrob Agents Chemother. 2011; 55:3538-45. http://www.ncbi.nlm.nih.gov/pubmed/21576437?dopt=AbstractPlus

718. Chisholm SA, Alexander S, Desouza-Thomas L et al. Emergence of a Neisseria gonorrhoeae clone showing decreased susceptibility to cefixime in England and Wales. J Antimicrob Chemother. 2011; :. http://www.ncbi.nlm.nih.gov/pubmed/21846672?dopt=AbstractPlus

719. Ison CA, Hussey J, Sankar KN et al. Gonorrhoea treatment failures to cefixime and azithromycin in England, 2010. Euro Surveill. 2011; 16 http://www.ncbi.nlm.nih.gov/pubmed/21492528?dopt=AbstractPlus

720. Unemo M, Golparian D, Hestner A. Ceftriaxone treatment failure of pharyngeal gonorrhoea verified by international recommendations, Sweden, July 2010. Euro Surveill. 2011; 16 http://www.ncbi.nlm.nih.gov/pubmed/21329645?dopt=AbstractPlus

721. Ohnishi M, Saika T, Hoshina S et al. Ceftriaxone-resistant Neisseria gonorrhoeae, Japan. Emerg Infect Dis. 2011; 17:148-9. http://www.ncbi.nlm.nih.gov/pubmed/21192886?dopt=AbstractPlus

722. Centers for Disease Control and Prevention (CDC). Update to CDC's Sexually transmitted diseases treatment guidelines, 2010: oral cephalosporins no longer a recommended treatment for gonococcal infections. MMWR Morb Mortal Wkly Rep. 2012; 61:590-4. http://www.ncbi.nlm.nih.gov/pubmed/22874837?dopt=AbstractPlus

723. Allen VG, Mitterni L, Seah C et al. Neisseria gonorrhoeae treatment failure and susceptibility to cefixime in Toronto, Canada. JAMA. 2013; 309:163-70. http://www.ncbi.nlm.nih.gov/pubmed/23299608?dopt=AbstractPlus

724. Kirkcaldy RD, Bolan GA, Wasserheit JN. Cephalosporin-resistant gonorrhea in North America. JAMA. 2013; 309:185-7. http://www.ncbi.nlm.nih.gov/pubmed/23299612?dopt=AbstractPlus

725. Olsen B, Pham TL, Golparian D et al. Antimicrobial susceptibility and genetic characteristics of Neisseria gonorrhoeae isolates from Vietnam, 2011. BMC Infect Dis. 2013; 13:40. http://www.ncbi.nlm.nih.gov/pubmed/23351067?dopt=AbstractPlus

726. Kirkcaldy RD, Zaidi A, Hook EW et al. Neisseria gonorrhoeae antimicrobial resistance among men who have sex with men and men who have sex exclusively with women: the Gonococcal Isolate Surveillance Project, 2005-2010. Ann Intern Med. 2013; 158:321-8. http://www.ncbi.nlm.nih.gov/pubmed/23460055?dopt=AbstractPlus

727. Goire N, Lahra MM, Ohnishi M et al. Polymerase chain reaction-based screening for the ceftriaxone-resistant Neisseria gonorrhoeae F89 strain. Euro Surveill. 2013; 18:20444. http://www.ncbi.nlm.nih.gov/pubmed/23594520?dopt=AbstractPlus

728. Forest Pharmaceuticals, Inc. Avycaz (ceftazidime and avibactam sodium) for injection prescribing information. Cincinnati, OH; 2015 Feb.

729. Cubist Pharmaceuticals, Inc. Zerbaxa (ceftolozane sulfate and tazobactam sodium) for injection prescribing information. Lexington, MA; 2015 May.