ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Gentamicin (systemic): Pediatric drug information

Gentamicin (systemic): Pediatric drug information
(For additional information see "Gentamicin (systemic): Drug information" and see "Gentamicin (systemic): Patient drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
ALERT: US Boxed Warning
Toxicity:

Ensure that patients treated with aminoglycosides are under close clinical observation because of the potential toxicity associated with their use.

As with other aminoglycosides, gentamicin is potentially nephrotoxic. The risk of nephrotoxicity is greater in patients with impaired renal function and in those who receive high dosages or prolonged therapy.

Neurotoxicity, manifested by ototoxicity, both vestibular and auditory, can occur in patients treated with gentamicin, primarily in those with preexisting renal damage and in patients with healthy renal function treated with higher doses and/or for longer periods than recommended. Aminoglycoside-induced ototoxicity is usually irreversible. Other manifestations of neurotoxicity may include numbness, skin tingling, muscle twitching, and convulsions.

Closely monitor renal and eighth cranial nerve functions, especially in patients with known or suspected reduced renal function at onset of therapy, and also in those whose renal function is initially normal but who develop signs of renal dysfunction during therapy. Examine urine for decreased specific gravity, increased excretion of protein, and the presence of cells or casts. Periodically determine serum urea nitrogen (BUN), serum creatinine, or creatinine clearance (CrCl). When feasible, it is recommended that serial audiograms be obtained in patients old enough to be tested, particularly high-risk patients. Evidence of ototoxicity (dizziness, vertigo, tinnitus, roaring in the ears, or hearing loss) or nephrotoxicity requires dosage adjustment or discontinuance of the drug. As with the other aminoglycosides, on rare occasions, changes in renal and eighth cranial nerve function may not manifest until soon after completion of therapy.

Monitor serum concentrations of aminoglycosides when feasible to ensure adequate levels and to avoid potentially toxic levels. When monitoring gentamicin peak concentrations, adjust dosage so that prolonged levels above 12 mcg/mL are avoided. When monitoring gentamicin trough concentrations, adjust dosage so that levels above 2 mcg/mL are avoided. Excessive peak or trough serum concentrations of aminoglycosides may increase the risk of renal and eighth cranial nerve toxicity. In the event of overdose or toxic reactions, hemodialysis may aid in the removal of gentamicin from the blood, especially if renal function is, or becomes, compromised. The rate of removal of gentamicin is considerably less by peritoneal dialysis than by hemodialysis.

Avoid concurrent and/or sequential systemic or topical use of other potentially neurotoxic and/or nephrotoxic drugs, such as cisplatin, cephaloridine, kanamycin, amikacin, neomycin, polymyxin B, colistin, paromomycin, streptomycin, tobramycin, vancomycin, and viomycin. Other factors that may increase patient risk of toxicity are advanced age and dehydration.

Avoid the concurrent use of gentamicin with potent diuretics, such as ethacrynic acid or furosemide, since certain diuretics by themselves may cause ototoxicity. In addition, when administered intravenously (IV), diuretics may enhance aminoglycoside toxicity by altering the antibiotic concentration in serum and tissue.

Pregnancy:

Aminoglycosides can cause fetal harm when administered to a pregnant woman.

Therapeutic Category
  • Antibiotic, Aminoglycoside
Dosing: Neonatal

Dosage guidance:

Dosing: Determination of dosing interval requires consideration of multiple factors including concomitant medications (eg, ibuprofen, indomethacin), history of birth depression, birth hypoxia/asphyxia, and presence of cyanotic congenital heart disease. Dosage should be individualized based upon serum concentration monitoring. Impact of extracorporeal membrane oxygenation (ECMO) on neonatal pharmacokinetic parameters is not fully elucidated; optimal dose and frequency not established in ECMO patients; available data are very limited (Ref). Patient-specific considerations (eg, reason for ECMO) and variability with ECMO procedure itself make extrapolation of pharmacokinetic data and dosing to all patients receiving ECMO difficult; closely monitor serum concentrations and determine individual dosing needs in these patients.

General dosing; susceptible infection: Limited data available: Dosing strategies may vary by institution as a wide variety of extended-interval dosing regimens have been studied (Ref). Due to maturational processes and developing pharmacokinetics, regimens referred to as "extended-interval" in neonates do not typically achieve concentrations that would be expected with extended-interval dosing in children and adults (Ref). Some dosing is based on tobramycin studies.

Age-directed dosing (Ref): IM, IV: Note: Higher doses and different dosing intervals may be required to achieve target concentrations if MIC ≥1 mg/L (Ref).

Gestational

Age

Postnatal

Age

Dose

<30 weeks

≤14 days

5 mg/kg/dose every 48 hours

≥15 days

5 mg/kg/dose every 36 hours

30 to 34 weeks

≤10 days

5 mg/kg/dose every 36 hours

11 to 60 days

5 mg/kg/dose every 24 hours

≥35 weeks

≤7 days

4 mg/kg/dose every 24 hours

8 to 60 days

5 mg/kg/dose every 24 hours

CNS infection (VP-shunt infection, ventriculitis): Limited data available: Intraventricular/intrathecal (use a preservative-free preparation): 1 mg/day (Ref). Note: A trial evaluating routine use of a higher dose (2.5 mg/day) reported a higher mortality rate in the intraventricular treatment group (Ref).

Dosing adjustment in renal impairment: Consider single-dose administration with serum concentration monitoring rather than scheduled dosing in patients with urine output <1 mL/kg/hour or if serum creatinine significantly increases from baseline.

Dosing: Pediatric

Dosage guidance:

Dosing: Initial dosing recommendations presented. Monitoring of serum concentrations is recommended to ensure efficacy and avoid toxicity, particularly in critically ill patients with serious infection or in disease states known to significantly alter aminoglycoside pharmacokinetics (eg, cystic fibrosis, burns, major surgery). Timing and frequency of concentration monitoring is individualized based on dosing and monitoring strategy (Ref). Routes of administration may vary (including IM, IV, intraperitoneal, intrathecal, and intraventricular); use caution. Some dosing is based on tobramycin studies.

Dosing consideration for obesity: In obese pediatric patients, use adjusted body weight (IBW + 0.4 [TBW – IBW]) to calculate initial dosage (Ref). Alternatively, adjusted body weight for obese pediatric patients may be calculated using the equation 0.7 x TBW (Ref), or fat-free mass can be used to calculate the initial dose in pediatric patients ≥2 years regardless of body habitus (Ref). Dosage should then be individualized based upon serum concentration monitoring.

General dosing, susceptible infection: Note: Optimal dose and frequency not established in patients receiving extracorporeal membrane oxygenation (ECMO); patient-specific considerations (eg, reason for ECMO) and variability with ECMO procedure itself make extrapolation of pharmacokinetic data and dosing to all patients receiving ECMO difficult; closely monitor serum concentrations and determine individual dosing needs in these patients.

Conventional dosing: Infants, Children, and Adolescents: IM, IV: 2 to 2.5 mg/kg/dose every 8 hours (Ref).

Extended-interval dosing: Limited data available:

Weight-directed: Infants, Children, and Adolescents: IV: 5 to 7.5 mg/kg/dose every 24 hours in patients with normal renal function (Ref).

Age-directed: Based on data from 114 patients, the following has been suggested (Ref):

Infants and Children ≥3 months to <2 years: IV: 9.5 mg/kg/dose every 24 hours.

Children 2 to <8 years: IV: 8.5 mg/kg/dose every 24 hours.

Children ≥8 years and Adolescents: IV: 7 mg/kg/dose every 24 hours.

Brucellosis

Brucellosis: Limited data available: Infants, Children, and Adolescents: IV, IM: 5 mg/kg/dose once daily as part of an appropriate combination regimen (Ref). Note: Duration depends on extent of disease; gentamicin is usually given for the first 7 to 14 days of therapy (Ref).

CNS infection

CNS infection:

Meningitis, including health care-associated meningitis: Infants, Children, and Adolescents: IV: 7.5 mg/kg/day divided every 8 hours in combination with other antibiotics; duration should be individualized based on patient characteristics, infecting organism, and response (Ref).

Ventriculitis, including health care–associated ventriculitis and cerebrospinal fluid (CSF) shunt infections: Limited data available; optimal dose not established (Ref). Dosage and administration interval can be adjusted based on CSF gentamicin concentrations, ventricle size, and daily output from ventricular drain. Duration is individualized according to clinical and microbiological response (Ref).

Intraventricular/intrathecal (use a preservative-free preparation):

Infants and Children: 1 to 2 mg/day.

Adolescents: Dosing recommendations not reported; however, in adults: 4 to 8 mg/day has been suggested.

Cystic fibrosis, pulmonary infection

Cystic fibrosis, pulmonary infection: Infants, Children, and Adolescents:

Conventional dosing: IM, IV: 3.3 mg/kg/dose every 8 hours (Ref).

Extended-interval dosing: IV: 10 to 12 mg/kg/dose every 24 hours (Ref); maximum reported dose from a survey of 28 Cystic Fibrosis (CF) Foundation-accredited centers ranged from 12 to 20 mg/kg/dose (Ref). Note: The CF Foundation recommends extended-interval dosing as preferred over conventional dosing.

Endocarditis, treatment

Endocarditis, treatment:

Synergy dosing (eg, gram-positive bacteria): Children and Adolescents: IV: 3 to 6 mg/kg/day divided every 8 hours; use in combination with other antibiotics dependent on pathogen and source of infection (ie, valve type) (Ref).

Treatment dosing (eg, gram-negative bacteria): Children and Adolescents: IV: 7.5 mg/kg/day divided every 8 hours; use in combination with other antibiotics (Ref).

Gonococcal infection, uncomplicated infection of the cervix, urethra, or rectum

Gonococcal infection, uncomplicated infection of the cervix, urethra, or rectum (alternative agent for severe cephalosporin allergy): Limited data available:

Children ≥45 kg and Adolescents: IM: 240 mg once (administer as two 3 mL injections) in combination with single dose of oral azithromycin (Ref). Note: For treatment failure, consult an infectious diseases specialist and report to the CDC through state and local health departments within 24 hours of diagnosis (Ref).

Intra-abdominal infection, complicated

Intra-abdominal infection, complicated: Infants, Children, and Adolescents: IV: 3 to 7.5 mg/kg/day divided every 8 to 24 hours; use in combination with other antibiotics (Ref).

Peritonitis

Peritonitis (peritoneal dialysis) (Ref): Limited data available: Infants, Children, and Adolescents:

Intermittent: Intraperitoneal:

Anuric: 0.6 mg/kg/dose every 24 hours in the long dwell.

Nonanuric: 0.75 mg/kg/dose every 24 hours in the long dwell.

Continuous: Intraperitoneal: Loading dose: 8 mg per liter of dialysate; maintenance dose: 4 mg per liter.

Surgical prophylaxis

Surgical prophylaxis: Infants, Children, and Adolescents: IV: 2.5 mg/kg as a single dose; administer within 60 minutes prior to surgical incision with or without other antibiotics (procedure dependent) (Ref).

Tularemia

Tularemia: Infants, Children, and Adolescents: IM, IV: 5 to 6 mg/kg/day in 2 to 3 divided doses (Ref); usual duration is 7 to 10 days; a shorter course (ie, 5 to 7 days) may be considered for mild disease; longer duration and concomitant antibiotics are required for severe illness (eg, meningitis) (Ref).

Urinary tract infection

Urinary tract infection (UTI):

Note: Duration of therapy for multiple-dose regimens: For uncomplicated cystitis in patients ≥3 months of age, treat for 3 to 5 days; patients <2 years of age may require a longer course (eg, 7 days). For complicated UTI, including pyelonephritis, treat for a total of 6 to 10 days; while 7 to 14 days has been recommended for complicated UTI, this was not shown to improve outcomes compared to a shorter duration of 6 to 10 days (Ref).

Conventional dosing: Infants, Children, and Adolescents: IV: 7.5 mg/kg/day divided every 8 hours until clinical improvement and able to tolerate oral intake; complete course with oral antibiotics. Duration should be individualized based upon age, severity, and degree of urinary tract involvement.

Extended-interval dosing: Limited data available: Based on data from 90 patients (ages: 1 month to 12 years), the following age-directed dosing has been suggested (Ref): Note: Patients were transitioned to oral therapy once afebrile for 24 hours.

Infants and Children <5 years: IV: 7.5 mg/kg/dose every 24 hours.

Children 5 to 10 years: IV: 6 mg/kg/dose every 24 hours.

Children 11 to 12 years: IV: 4.5 mg/kg/dose every 24 hours.

Single-dose regimen: Limited data available: Note: Recommended for treatment of uncomplicated cystitis caused by antimicrobial resistant gram-negative pathogens (Ref):

Infants, Children, and Adolescents: IM: 5 mg/kg as a single dose; dosing based on 2 prospective studies evaluating single-dose IM gentamicin in patients 1 month to 15 years and a systematic review evaluating studies of various single-dose aminoglycosides in pediatric and adult patients (Ref); one study limited doses to 300 mg (Ref). An overall pooled cure rate for single dose IM aminoglycoside for treatment of children and adults with mainly uncomplicated cystitis was reported as 94.5% ± 4.3% (Ref). Note: Guidelines do not address pediatric dosing; recommended aminoglycoside doses in adults include IV single doses (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Pediatric

Parenteral: Note: Gentamicin serum concentrations should be monitored in patients with kidney impairment; following the initial dose, subsequent doses may be determined based on therapeutic monitoring.

Infants, Children, and Adolescents: IM, IV:

The following adjustments have been recommended (Ref): Note: Renally adjusted dose recommendations are based on doses of 2.5 mg/kg/dose every 8 hours:

GFR >50 mL/minute/1.73 m2: No dosage adjustment necessary.

GFR 30 to 50 mL/minute/1.73 m2: Administer every 12 to 18 hours.

GFR 10 to 29 mL/minute/1.73 m2: Administer every 18 to 24 hours.

GFR <10 mL/minute/1.73 m2: Administer every 48 to 72 hours.

Intermittent hemodialysis: 2 mg/kg/dose; redose as indicated by serum concentration.

Peritoneal dialysis (PD): 2 mg/kg/dose; redose as indicated by serum concentration.

Continuous renal replacement therapy (CRRT): 2 to 2.5 mg/kg/dose every 12 to 24 hours, monitor serum concentrations.

Dosing: Hepatic Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling; however, dosage adjustment not likely to be necessary (does not undergo hepatic metabolism).

Dosing: Adult

(For additional information see "Gentamicin (systemic): Drug information")

Dosage guidance:

Dosing: For patients who are underweight (ie, total body weight [TBW] < ideal body weight [IBW]), calculate the dose based on TBW. For patients who are nonobese (ie, TBW 1 to 1.25 × IBW), calculate the dose based on TBW or IBW. TBW may be preferred in patients who are nonobese who may have increased Vd (eg, critically ill). For patients with obesity (ie, TBW >1.25 x IBW), use adjusted body weight ([0.4 x {TBW-IBW}] + IBW) for initial weight-based dosing and for estimating kidney function with Cockcroft-Gault (CrCl) (Ref).

Clinical considerations: Therapeutic drug monitoring is recommended to ensure efficacy and avoid toxicity, particularly in patients who are critically ill with serious infections or in disease states known to significantly alter aminoglycoside pharmacokinetics (eg, cystic fibrosis, burns, major surgery). Timing and frequency of concentration monitoring is individualized based on dosing and monitoring strategy (Ref).

Usual dosage range:

Gram-negative infections:

Conventional/traditional dosing: IV, IM: 3 to 5 mg/kg/day in divided doses every 8 hours (Ref). Some experts favor an initial loading dose of 2.5 to 3 mg/kg (Ref). Target peak concentration depends on indication and site of infection; in general, adjust dose to achieve peak of 4 to 6 mcg/mL for urinary tract infections and 7 to 10 mcg/mL for serious infections (including life-threatening infections). Target trough concentrations should be <2 mcg/mL; ideal target <1 mcg/mL (Ref).

High-dose extended-interval dosing (once-daily dosing): IV: 5 to 7 mg/kg once daily; use with caution in patients with CrCl <40 mL/minute (Ref). Adjust gentamicin dose and interval to achieve an extrapolated peak concentration of ~15 to 20 mcg/mL and trough concentration ≤0.5 mcg/mL (Ref). Note: Published nomograms for dosage adjustment may not apply to patients with altered pharmacokinetics (eg, patients with ascites, burns covering >20% total BSA, end-stage renal disease requiring hemodialysis, pregnancy) (Ref); some experts prefer traditional intermittent dosing in such populations (Ref).

Synergy dosing for non-CNS gram-positive infections:

IV, IM: 3 mg/kg/day in 1 to 3 divided doses in combination with a gram-positive active agent (Ref). When divided doses are used, adjust gentamicin dose to achieve peak concentration of 3 to 4 mcg/mL and trough concentration <1 mcg/mL (Ref).

Bartonella spp. infection

Bartonella spp. infection (off-label use):

Bacteremia with or without endocarditis: IV: 3 mg/kg/day in 1 or 3 divided doses in combination with doxycycline for 2 weeks, followed by doxycycline monotherapy (Ref). Some experts do not suggest gentamicin as part of the regimen for patients with HIV and bacteremia without endocarditis (Ref).

Cat scratch disease, disseminated (eg, hepatosplenic, prolonged systemic febrile illness) (alternative agent): IV: 3 mg/kg once daily in combination with rifampin for 10 to 14 days (Ref).

Note: In contrast to other gram-negative infections, target a gentamicin peak concentration of 3 to 4 mcg/mL and trough concentration of <1 mcg/mL when divided doses are used (Ref).

Bloodstream infection

Bloodstream infection:

Adjunctive empiric therapy for patients with sepsis/septic shock and concern for resistant gram-negative bacteria (eg, immunosuppression, prevalent local resistance, recent antibiotic exposure): IV: 5 to 7 mg/kg once daily in combination with a second gram-negative active agent; once culture and susceptibility results are available, can generally discontinue and use a single agent with documented activity. Gentamicin should not be used as monotherapy (Ref).

Antibiotic lock technique (catheter-salvage strategy) (off-label use): Note: For infections caused by susceptible organisms when the catheter cannot be removed; use in addition to systemic antibiotics (Ref).

Intracatheter: Prepare lock solution to final concentration of gentamicin 1 to 5 mg/mL (may be combined with heparin) (Ref). The gentamicin concentration may vary by institution, catheter type, and whether heparin is utilized; solutions with heparin are preferred. Prepare solution immediately before instillation with a sufficient volume to fill the catheter (2 to 5 mL). Instill into each lumen of the catheter access port with a dwell time of up to 72 hours, depending on frequency of catheter use. Withdraw lock solution prior to catheter use; replace with fresh gentamicin lock solution after catheter use. Antibiotic lock therapy is given for the same duration as systemic antibiotics (Ref).

Brucellosis

Brucellosis (off-label use):

Note: Additional agents or other regimens are preferred for neurobrucellosis and infection in patients who are pregnant (Ref).

Endocarditis: IV, IM: 5 mg/kg/day in 1 to 3 divided doses for 4 weeks as part of an appropriate combination regimen (Ref).

Spondylitis: IV, IM: 5 mg/kg once daily for 7 to 14 days as part of an appropriate combination regimen (Ref).

Uncomplicated (nonfocal): IV, IM: 5 mg/kg once daily for 7 to 10 days as part of an appropriate combination regimen (Ref).

CNS infection, health care–associated

CNS infection, health care–associated (eg, cerebrospinal fluid [CSF] shunt infection) (adjunct to systemic therapy):

Note: Reserved for infections due to multidrug-resistant organisms, infections refractory to appropriate parenteral therapy, or when infected shunts cannot be removed (Ref).

Intraventricular (use a preservative-free preparation): Initial: 4 to 8 mg/day; some experts recommend adjusting dosage and administration interval based on CSF gentamicin concentrations (goal: 10 to 20 times MIC of causative organism), ventricle size, and daily output from ventricular drain (Ref). When intraventricular gentamicin is administered via a ventricular drain, clamp drain for 15 to 60 minutes after administration (allows solution to equilibrate in CSF). Duration is individualized according to clinical and microbiologic response (Ref).

Endocarditis, treatment

Endocarditis, treatment:

Enterococcus spp. (native or prosthetic valve, without high-level gentamicin resistance): IV, IM: 1 mg/kg every 8 hours as part of an appropriate combination regimen. Duration is 4 to 6 weeks depending on duration of symptoms prior to presentation, source of infection, and the specific combination regimen (Ref). Note: For native-valve endocarditis due to ampicillin-susceptible Enterococcus faecalis, some experts prefer a combination regimen that does not contain gentamicin (Ref).

Staphylococcus spp. (prosthetic valve) (off-label use): IV, IM: 1 mg/kg every 8 hours for the first 2 weeks of an appropriate combination regimen; the other components should be continued for a total of ≥6 weeks (Ref).

Viridans group streptococci and Streptococcus bovis (off-label use):

Native valve: Highly penicillin-susceptible (MIC ≤0.12 mcg/mL): IV, IM: 3 mg/kg/day once daily (preferred) or in 3 divided doses (alternative) in combination with a beta-lactam for 2 weeks. Note: This regimen is reserved for patients with uncomplicated infection, prompt response to therapy, and no preexisting renal failure (Ref).

Native valve: Relatively penicillin-resistant (MIC >0.12 and <0.5 mcg/mL): IV, IM: 3 mg/kg/day once daily (preferred) or in 3 divided doses (alternative) in combination with a beta-lactam. Gentamicin duration is for the first 2 weeks of the total 4-week regimen (Ref).

Native valve: Penicillin-resistant (MIC ≥0.5 mcg/mL): IV, IM: 3 mg/kg/day in 2 to 3 divided doses in combination with a beta-lactam. The duration of this regimen is not well established; infectious diseases consultation recommended (Ref).

Prosthetic valve: Highly penicillin-susceptible (MIC ≤0.12 mcg/mL): IV, IM: 3 mg/kg/day once daily (preferred) or in 3 divided doses (alternative) in combination with a beta-lactam. Gentamicin duration is for the first 2 weeks of the total 6-week regimen (Ref).

Prosthetic valve: Relatively penicillin-resistant (MIC >0.12 and <0.5 mcg/mL) or fully penicillin-resistant (MIC ≥0.5 mcg/mL): IV, IM: 3 mg/kg/day once daily or in 3 divided doses in combination with a beta-lactam for 6 weeks (Ref); some experts prefer 3 divided doses for these isolates and also prefer shorter courses of the gentamicin component (≥2 weeks) for relatively penicillin-resistant strains (Ref).

Meningitis, bacterial

Meningitis, bacterial:

Enterococcus spp.: IV: 5 mg/kg/day in 1 or 3 divided doses (Ref); give as part of an appropriate combination regimen and individualize duration based on clinical response (Ref).

Listeria monocytogenes: IV: 5 mg/kg/day in 3 divided doses in combination with ampicillin or penicillin. Gentamicin is given until clinical improvement (typically ≥7 days) or in poor responders for up to 21 days, as long as there are no signs of nephrotoxicity or ototoxicity; total duration of antibiotic therapy is ≥21 days (Ref).

Osteomyelitis, prevention, following open fractures

Osteomyelitis, prevention, following open fractures (type III [severe contamination or comminution]) (off-label use): IV: 5 mg/kg every 24 hours as part of an appropriate combination regimen. Duration is 72 hours after injury or up to 24 hours after wound closure (Ref).

Pelvic infections

Pelvic infections (off-label use):

Note: Dose is based on actual body weight. For patients >1.2 × IBW, use adjusted body weight (IBW + [0.4 × (TBW-IBW)]) (Ref).

Intra-amniotic infection (chorioamnionitis): IV: 5 mg/kg once daily (preferred) or 2 mg/kg loading dose followed by 1.5 mg/kg every 8 hours (alternative); give in combination with ampicillin. In women undergoing cesarean delivery, an anti-anaerobic agent should also be added. Continue regimen until vaginal delivery or for 1 dose after cesarean delivery (Ref). Note: Some experts recommend 1 additional dose after vaginal delivery and extension of antibiotics after cesarean delivery until patient is afebrile and asymptomatic ≥48 hours (Ref).

Postpartum endometritis: IV: 5 mg/kg once daily (preferred) or 1.5 mg/kg every 8 hours (alternative) in combination with clindamycin. Treat until the patient is clinically improved (no fundal tenderness) and afebrile for 24 to 48 hours (Ref). Note: For women with group B Streptococcus colonization or sepsis, additional agents or other regimens are preferred (Ref).

Peritonitis, treatment

Peritonitis, treatment (peritoneal dialysis patients) (off-label use): As a component of empiric therapy or for pathogen-directed therapy.

Note: Intraperitoneal administration is preferred to IV administration. Once culture results are available, switch to another active antibiotic class, if possible, to decrease the risk of toxicity; otherwise, duration of therapy is ≥3 weeks for patients with adequate clinical response (Ref). Consider a 25% dose increase in patients with significant residual renal function (urine output >100 mL/day) (Ref).

Intermittent (strongly preferred): Intraperitoneal: 0.6 mg/kg added to one exchange of dialysis solution once daily (allow to dwell ≥6 hours) (Ref).

Continuous (with every exchange) (dose is per liter of dialysate): Intraperitoneal: Loading dose: 8 mg/L of dialysate with first exchange of dialysate; maintenance dose: 4 mg/L of dialysate with each subsequent exchange of dialysate (Ref).

Plague

Plague ( Yersinia pestis ), treatment (off-label use):

Note: Consult public health officials for event-specific recommendations.

IV, IM: 5 mg/kg once daily for 7 to 14 days and for at least a few days after clinical resolution (Ref).

Sepsis or septic shock, adjunctive empiric gram-negative coverage

Sepsis or septic shock, adjunctive empiric gram-negative coverage (eg, in the setting of intra-abdominal infection, pneumonia, gram-negative bacteremia, or severe burn):

Note: Some experts reserve for patients with immunocompromising conditions or risk for resistant gram-negative pathogens, in particular Pseudomonas aeruginosa (Ref).

IV: 5 to 7 mg/kg once daily in combination with a second gram-negative agent (Ref); once culture and susceptibility tests are available, can generally discontinue and use a single agent with documented activity. Gentamicin should not be used as monotherapy for severe infections outside of the urinary tract (Ref).

Sexually transmitted infections

Sexually transmitted infections:

Gonococcal infection, uncomplicated (infection of the cervix, rectum, or urethra) (alternative agent) (off-label use):

Note: Reserve for patients who cannot use a cephalosporin (Ref).

>45 kg: IM: 240 mg (administer as two 3 mL injections) once (Ref).

≤45 kg: IM: 5 mg/kg once (Ref).

Administer in combination with oral azithromycin 2 g, each as a single dose (Ref). When treatment failure is suspected (eg, detection of Neisseria gonorrhoeae after treatment without additional sexual exposure), consult an infectious diseases specialist. Report failures to the CDC through state and local health departments (Ref).

Pelvic inflammatory disease, severe (including tubo-ovarian abscess) (alternative agent) (off-label use): IV, IM: 3 to 5 mg/kg once daily or 2 mg/kg as a loading dose followed by 1.5 mg/kg every 8 hours; give as part of an appropriate combination regimen; after 24 to 48 hours of sustained clinical improvement, transition to oral therapy to complete 14 days of treatment (Ref). Note: Some experts reserve this regimen for patients who cannot use preferred agents due to greater associated adverse effects (Ref).

Surgical prophylaxis

Surgical prophylaxis (alternative agent for select GI tract, GU tract, or gynecologic/obstetric procedures) (off-label use):

IV: 5 mg/kg as a single dose within 60 minutes prior to surgical incision; give in combination with other antibiotics (procedure dependent) (Ref). Note: In cases where extension of prophylaxis is warranted postoperatively, total duration should be ≤24 hours (Ref). Postoperative prophylaxis is not recommended in clean and clean-contaminated surgeries (Ref).

Tularemia

Tularemia (off-label use):

IV, IM: 5 mg/kg/day in 1 to 3 divided doses (Ref); duration is generally 7 to 10 days but should be extended for severe cases or for patients with a delayed response to treatment (Ref).

Urinary tract infection, complicated

Urinary tract infection, complicated (pyelonephritis or urinary tract infection with systemic signs/symptoms) (alternative agent): Note: Some experts reserve for use when other long-acting parenteral antimicrobials (eg, ceftriaxone) or fluoroquinolones cannot be used due to allergy, intolerance, unmodifiable drug interactions, or resistance (Ref).

Inpatients: IV, IM: 5 mg/kg once daily. Switch to an appropriate oral regimen once symptoms improve, if culture and susceptibility results allow. Total duration of therapy ranges from 5 to 14 days and depends on clinical response and the antimicrobial chosen to complete the regimen (Ref).

Outpatients: IV, IM: 5 mg/kg once, followed by 5 to 14 days of appropriate oral therapy (Ref). Note: For patients who are systemically ill or at risk for more severe illness, some experts continue daily parenteral therapy pending culture and susceptibility testing results (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Altered kidney function:

High-dose, extended-interval dosing:

Note: Use with caution in patients with CrCl <40 mL/minute (Ref); high-dose, extended-interval dosing may still be considered, especially in patients with severe sepsis/shock or those infected with multidrug-resistant gram-negative organisms (Ref).

IV: Initial dose: 5 to 7 mg/kg. Subsequent doses and frequency of administration should be determined based on therapeutic drug monitoring; regimens may vary; nomograms exist to guide dose adjustments, although individualized calculations may be necessary in patients with highly variable or altered aminoglycoside pharmacokinetics (eg, critical illness, pregnancy) (Ref). Also refer to institutional-specific policies. The following recommendations may serve as a general guideline after the initial dose:

CrCl ≥60 mL/minute: IV: Administer every 24 hours; adjust dose and/or interval based on gentamicin serum concentrations.

CrCl 40 to <60 mL/minute: IV: Administer every 36 hours; adjust dose and/or interval based on gentamicin serum concentrations.

CrCl 20 to <40 mL/minute: IV: Administer every 48 hours; adjust dose and/or interval based on gentamicin serum concentrations.

CrCl <20 mL/minute: IV: Administer usual dose once, then determine subsequent dose and interval based on serum concentration monitoring. Some published protocols would recommend conventional/traditional dosing in these patients (Ref).

Conventional/traditional dosing:

Note: High-dose, extended-interval dosing is generally preferred for treatment of gram-negative infections. Conventional/traditional dosing is typically used for synergy dosing or non-CNS, gram-positive infections.

Regimens may vary based on individualized pharmacokinetic calculations and pharmacodynamic targets; also refer to institutional-specific policies. Note: The following recommendations are expert opinion derived from Leroy 1978 and based on a usual dosage range of 3 to 5 mg/kg/day:

CrCl ≥60 mL/minute: IM, IV: No dosage adjustment necessary.

CrCl ≥40 to <60 mL/minute: IM, IV: Administer usual dose every 12 hours; adjust dose and/or interval based on gentamicin serum concentrations.

CrCl 20 to <40 mL/minute: IM, IV: Administer usual dose every 24 hours; adjust dose and/or interval based on gentamicin serum concentrations.

CrCl <20 mL/minute: IM, IV: Administer usual dose every 36 to 48 hours; adjust dose and/or interval based on gentamicin serum concentrations.

Augmented renal clearance (measured urinary CrCl ≥130 mL/minute/1.73 m2):

Note: Augmented renal clearance (ARC) is a condition that occurs in certain critically ill patients without organ dysfunction and with normal serum creatinine concentrations. Young patients (<55 years of age) admitted post trauma or major surgery are at highest risk for ARC, as well as those with sepsis, burns, or hematologic malignancies. An 8- to 24-hour measured urinary CrCl is necessary to identify these patients (Ref).

High-dose, extended-interval dosing: IV: Initial: 7 to 8 mg/kg once daily for known/suspected sepsis; adjust dose and/or interval based on close monitoring of gentamicin serum concentrations and individualized pharmacokinetic calculations and pharmacodynamic targets; use of nomograms for dosage adjustment are not recommended in this population (Ref).

Hemodialysis, intermittent (thrice weekly): Dialyzable (~50%; dependent on filter and duration): Note: Postdialysis concentrations should be drawn ≥2 and up to 4 hours after hemodialysis (HD) to allow for redistribution (Ref).

Loading dose of 2 to 3 mg/kg followed by:

Gram-positive synergy dosing: IM, IV: 1 mg/kg/dose 3 times weekly after dialysis on dialysis days (Ref); consider redosing for pre-HD or post-HD concentrations <1 mg/L.

Urinary tract infection: IM, IV: 1 to 1.5 mg/kg/dose 3 times weekly after dialysis on dialysis days (Ref); consider redosing for pre-HD concentrations <1.5 to 2 mg/L or post-HD concentrations <1 mg/L.

Systemic gram-negative infection: IM, IV: 2 to 3 mg/kg/dose 3 times weekly after dialysis on dialysis days (Ref); consider redosing for pre-HD concentrations <3 to 5 mg/L or post-HD concentrations <2 mg/L.

Peritoneal dialysis: IM, IV: Initial: 1 to 3 mg/kg/dose (depending on infection site, severity, and susceptibility of infecting organisms) every 48 to 72 hours based on gentamicin serum concentrations (Ref).

CRRT:

Note: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Recommendations are based on high-flux dialyzers and effluent flow rates of 20 to 25 mL/kg/hour (or ~1,500 to 3,000 mL/hour) unless otherwise noted. Appropriate dosing requires consideration of adequate drug concentrations (eg, site of infection) and consideration of initial loading doses. Close monitoring of response and adverse reactions (eg, nephrotoxicity) due to drug accumulation is important.

CVVH/CVVHD/CVVHDF:

Gram-positive synergy dosing: IV: Loading dose of 2 to 3 mg/kg/dose followed by 1 to 1.5 mg/kg every 24 to 36 hours (redose when concentration <1 mg/L (Ref)).

Urinary tract infection: IV: Loading dose of 2 to 3 mg/kg/dose followed by 1 to 1.5 mg/kg every 24 to 36 hours (redose when concentration <1.5 to 2 mg/L (Ref)).

Systemic gram-negative infection: IV: Initial: 2.5 to 3 mg/kg/dose; up to 5 mg/kg/dose may be considered (depending on infection site, severity, and susceptibility of infecting organisms) every 24 to 48 hours based on gentamicin serum concentrations (expert opinion derived from Petejova 2012).

PIRRT (eg, sustained, low-efficiency diafiltration):

Note: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Appropriate dosing requires consideration of adequate drug concentrations (eg, site of infection) and consideration of initial loading doses. Close monitoring of response and adverse reactions (eg, nephrotoxicity) due to drug accumulation is important. Dosing below assumes daily use of PIRRT.

Gram-positive synergy dosing: IV: Loading dose of 2 to 3 mg/kg/dose followed by 1 to 1.5 mg/kg/dose every 24 to 36 hours; adjust dose and/or frequency based on gentamicin serum concentrations. When scheduled dose falls on a PIRRT day, administer after PIRRT (Ref).

Urinary tract infection: IV: Loading dose of 2 to 3 mg/kg/dose followed by 1 to 1.5 mg/kg/dose every 24 to 36 hours; adjust dose based on serum concentrations. When scheduled dose falls on a PIRRT day, administer after PIRRT (Ref).

Systemic gram-negative infection: IV: Initial: 6 mg/kg/dose every 48 hours; adjust interval if needed based on gentamicin levels (usual frequency is every 48 to 72 hours). Note: Administer each dose 30 to 60 minutes prior to PIRRT session on PIRRT days (Ref).

Dosing: Hepatic Impairment: Adult

There are no dosage adjustments provided in the manufacturer’s labeling; however dosage adjustment is not likely to be necessary (does not undergo hepatic metabolism).

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified. Frequency not defined.

Cardiovascular: Edema, hypertension, hypotension, phlebitis, thrombophlebitis

Central nervous system: Abnormal gait, ataxia, brain disease, confusion, depression, dizziness, drowsiness, headache, lethargy, myasthenia, numbness, paresthesia, peripheral neuropathy, pseudomotor cerebri, seizure, vertigo

Dermatologic: Alopecia, erythema, pruritus, skin rash, urticaria

Endocrine & metabolic: Hypocalcemia, hypokalemia, hypomagnesemia, hyponatremia, weight loss

Gastrointestinal: Anorexia, Clostridioides difficile-associated diarrhea, decreased appetite, enterocolitis, nausea, sialorrhea, stomatitis, vomiting

Genitourinary: Casts in urine (hyaline, granular), Fanconi-like syndrome (infants and adults; high dose, prolonged course), oliguria, proteinuria

Hematologic & oncologic: Agranulocytosis, anemia, eosinophilia, granulocytopenia, leukopenia, purpura, reticulocytopenia, reticulocytosis, splenomegaly, thrombocytopenia

Hepatic: Hepatomegaly, increased liver enzymes

Hypersensitivity: Anaphylaxis, anaphylactoid reaction, hypersensitivity reaction

Local: Injection site reaction, pain at injection site

Neuromuscular & skeletal: Arthralgia, muscle cramps, muscle fatigue (myasthenia gravis-like syndrome), muscle twitching, tremor, weakness

Ophthalmic: Visual disturbance

Otic: Auditory impairment, hearing loss (associated with persistently increased serum concentrations; early toxicity usually affects high-pitched sound), tinnitus

Renal: Decreased creatinine clearance, decreased urine specific gravity, increased blood urea nitrogen, increased serum creatinine, polyuria, renal failure (high trough serum concentrations), renal tubular necrosis

Respiratory: Dyspnea, laryngeal edema, pulmonary fibrosis, respiratory depression

Miscellaneous: Fever

Contraindications

Hypersensitivity to gentamicin, other aminoglycosides, or any component of the formulation

Warnings/Precautions

Concerns related to adverse effects:

• Hypersensitivity: Cross-sensitivity to other aminoglycosides may occur.

• Nephrotoxicity: [US Boxed Warning]: May cause nephrotoxicity; usual risk factors include preexisting renal impairment, concomitant nephrotoxic medications, advanced age and dehydration. Discontinue treatment if signs of nephrotoxicity occur; renal damage is usually reversible.

• Neuromuscular blockade and respiratory paralysis: May cause neuromuscular blockade and respiratory paralysis; especially when given soon after anesthesia or neuromuscular blockers.

• Neurotoxicity: [US Boxed Warning]: May cause neurotoxicity; usual risk factors include preexisting renal impairment, concomitant neuro-/nephrotoxic medications, advanced age and dehydration. Ototoxicity is proportional to the amount of drug given and the duration of treatment. Tinnitus or vertigo may be indications of vestibular injury and impending bilateral irreversible damage. Discontinue treatment if signs of ototoxicity occur.

• Superinfection: Prolonged use may result in fungal or bacterial superinfection, including Clostridioides difficile-associated diarrhea (CDAD) and pseudomembranous colitis; CDAD has been observed >2 months postantibiotic treatment.

Disease-related concerns:

• Electrolyte abnormalities: Use with caution in patients with hypocalcemia, hypokalemia, or hypomagnesemia.

• Hearing impairment: Use with caution in patients with preexisting vertigo, tinnitus, or hearing loss.

• Neuromuscular disorders: Use with caution in patients with neuromuscular disorders, including myasthenia gravis.

• Renal impairment: Use with caution in patients with preexisting renal insufficiency; dosage modification required.

Special populations:

• Patients with genomic variants in MT-RNR1: Carriers of certain variants in the MT-RNR1 gene (eg, m.1555A>G) may be at increased risk for aminoglycoside-induced ototoxicity, including potentially significant hearing loss that may be irreversible, even when serum levels are within the normal range.

• Pregnancy: [US Boxed Warning]: Aminoglycosides may cause fetal harm if administered to a pregnant woman.

Concurrent drug therapy issues:

• Neurotoxic and/or nephrotoxic drugs: [US Boxed Warning]: Avoid concomitant or sequential use of other neurotoxic and/or nephrotoxic drugs (eg, cisplatin, polymyxin B, colistin, vancomycin, other aminoglycosides).

• Potent diuretics: [US Boxed Warning]: Avoid concomitant use with potent diuretics (eg, ethacrynic acid, furosemide) since diuretics themselves may cause ototoxicity and may enhance aminoglycoside toxicity.

Other warnings/precautions:

• Long-term use: Risk of toxicity is increased with extended duration of administration; additional monitoring may be required with long-term use.

• Surgical irrigation: May be almost completely systemically absorbed after local irrigation and/or topical application (except to the urinary bladder) during surgical procedures. Consider potential for nephrotoxicity, neuromuscular blockade, ototoxicity, and respiratory paralysis when administering aminoglycosides in this manner.

Warnings: Additional Pediatric Considerations

Use with caution in pediatric patients on extracorporeal membrane oxygenation (ECMO); pharmacokinetics of aminoglycosides may be altered; dosage adjustment and close monitoring necessary. Oral use for the prevention of NEC in premature neonates may potentially increase the risk for development of resistant bacteria; routine use for this is not recommended (Bury 2001; Reber 2004).

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Generic: 40 mg/mL (2 mL, 20 mL)

Solution, Injection [preservative free]:

Generic: 10 mg/mL (2 mL)

Solution, Intravenous [preservative free]:

Generic: 60 mg (50 mL); 80 mg (50 mL, 100 mL); 100 mg (50 mL, 100 mL); 120 mg (100 mL)

Generic Equivalent Available: US

Yes

Pricing: US

Solution (Gentamicin Sulfate Injection)

10 mg/mL (per mL): $1.03 - $3.25

40 mg/mL (per mL): $0.73 - $2.44

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Generic: 10 mg/mL (2 mL); 40 mg/mL (2 mL, 20 mL)

Solution, Intravenous:

Generic: 80 mg (50 mL); 100 mg (100 mL)

Administration: Pediatric

Parenteral:

IM: May be administered undiluted.

IV: Administer as diluted solution by slow intermittent infusion over 30 to 120 minutes; usual infusion time is 30 to 60 minutes; consider longer infusion time (60 to 120 minutes) with high doses. Shorter infusion times (≤5 minutes) have been reported in pediatric patients, including preterm and term neonates, receiving ≤4 mg/kg/dose (Ref). Avoid infusing concomitantly with penicillins or cephalosporins if feasible; consult drug interactions database for more information.

Intrathecal/intraventricular: Use preservative-free preparations only; must be diluted prior to administration. No specific administration information available; it has been suggested that instillation of small volumes (<3 mL) over 1 to 2 minutes is safe (Ref). When administered through a ventricular drain, clamp drain for 15 to 60 minutes to allow gentamicin solution to equilibrate in the cerebrospinal fluid (CSF) (Ref).

Administration: Adult

IM: Administer undiluted. Gentamicin in NS is not intended for IM administration.

IV: Infuse over 30 to 120 minutes.

Some penicillins (eg, carbenicillin, ticarcillin, piperacillin) have been shown to inactivate aminoglycosides in vitro. This has been observed to a greater extent with tobramycin and gentamicin, while amikacin has shown greater stability against inactivation. Concurrent use of these agents may pose a risk of reduced antibacterial efficacy in vivo, particularly in the setting of profound renal impairment. However, definitive clinical evidence is lacking. If combination penicillin/aminoglycoside therapy is desired in a patient with renal dysfunction, separation of doses (if feasible), and routine monitoring of aminoglycoside levels, CBC, and clinical response should be considered.

Intraventricular (off-label route): Use preservative-free preparations only. When administered through a ventricular drain, clamp drain for 15 to 60 minutes before opening the drain to allow gentamicin solution to equilibrate in the cerebrospinal fluid (Ref).

Storage/Stability

Store intact vials and premixed bags at 20°C to 25°C (68°F to 77°F). Protect from freezing. IV infusion solutions mixed in NS or D5W are stable for 48 hours at room temperature and refrigeration (Goodwin 1991).

Use

Treatment of serious infections (eg, sepsis, meningitis, urinary tract infections, respiratory tract infections, peritonitis, bone infections, skin and soft tissue infections) caused by susceptible strains of the following microorganisms: Pseudomonas aeruginosa, Escherichia coli, Klebsiella, Proteus, Serratia, Enterobacter, Citrobacter, and Staphylococcus species (FDA approved in all ages); empiric treatment of sepsis, in combination with a penicillin antibiotic such as ampicillin (FDA approved in neonates); has also been used for the treatment of brucellosis, peritonitis in patients with peritoneal catheters, and tularemia and for prophylaxis of surgical site infections.

Medication Safety Issues
Sound-alike/look-alike issues:

Gentamicin may be confused with gentian violet, kanamycin, vancomycin

High alert medication:

The Institute for Safe Medication Practices (ISMP) includes this medication (intrathecal administration) among its list of drug classes which have a heightened risk of causing significant patient harm when used in error.

Metabolism/Transport Effects

None known.

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

Agalsidase Alfa: Gentamicin (Systemic) may diminish the therapeutic effect of Agalsidase Alfa. Risk X: Avoid combination

Agalsidase Beta: Gentamicin (Systemic) may diminish the therapeutic effect of Agalsidase Beta. Management: Avoid concomitant use of gentamicin with agalsidase beta when possible as gentamicin could antagonize intracellular alpha-galactosidase activity. Risk D: Consider therapy modification

Aminoglycosides: May enhance the nephrotoxic effect of other Aminoglycosides. Aminoglycosides may enhance the neurotoxic effect of other Aminoglycosides. Risk X: Avoid combination

Amphotericin B: May enhance the nephrotoxic effect of Aminoglycosides. Amphotericin B may enhance the neurotoxic effect of Aminoglycosides. Risk C: Monitor therapy

Ataluren: May enhance the adverse/toxic effect of Aminoglycosides. Specifically, an increased risk of nephrotoxicity may occur with the concomitant use of ataluren and aminoglycosides. Risk X: Avoid combination

Bacillus clausii: Antibiotics may diminish the therapeutic effect of Bacillus clausii. Management: Bacillus clausii should be taken in between antibiotic doses during concomitant therapy. Risk D: Consider therapy modification

Bacitracin (Systemic): May enhance the nephrotoxic effect of Aminoglycosides. Bacitracin (Systemic) may enhance the neurotoxic effect of Aminoglycosides. Risk X: Avoid combination

BCG (Intravesical): Antibiotics may diminish the therapeutic effect of BCG (Intravesical). Risk X: Avoid combination

BCG Vaccine (Immunization): Antibiotics may diminish the therapeutic effect of BCG Vaccine (Immunization). Risk C: Monitor therapy

Bisphosphonate Derivatives: Aminoglycosides may enhance the hypocalcemic effect of Bisphosphonate Derivatives. Aminoglycosides may enhance the nephrotoxic effect of Bisphosphonate Derivatives. Risk C: Monitor therapy

Botulinum Toxin-Containing Products: Aminoglycosides may enhance the neuromuscular-blocking effect of Botulinum Toxin-Containing Products. Risk C: Monitor therapy

Capreomycin: May enhance the neuromuscular-blocking effect of Aminoglycosides. Risk C: Monitor therapy

CARBOplatin: May enhance the nephrotoxic effect of Aminoglycosides. Aminoglycosides may enhance the ototoxic effect of CARBOplatin. Especially with higher doses of carboplatin. Risk C: Monitor therapy

Cardiac Glycosides: Aminoglycosides may decrease the serum concentration of Cardiac Glycosides. This effect has only been demonstrated with oral aminoglycoside administration. Risk C: Monitor therapy

Cephalosporins: May enhance the nephrotoxic effect of Aminoglycosides. Cephalosporins may decrease the serum concentration of Aminoglycosides. Risk C: Monitor therapy

Cholera Vaccine: Antibiotics may diminish the therapeutic effect of Cholera Vaccine. Management: Avoid cholera vaccine in patients receiving systemic antibiotics, and within 14 days following the use of oral or parenteral antibiotics. Risk X: Avoid combination

CISplatin: May enhance the nephrotoxic effect of Aminoglycosides. CISplatin may enhance the neurotoxic effect of Aminoglycosides. Risk X: Avoid combination

Colistimethate: Aminoglycosides may enhance the nephrotoxic effect of Colistimethate. Aminoglycosides may enhance the neuromuscular-blocking effect of Colistimethate. Management: Avoid coadministration of colistimethate and aminoglycosides whenever possible due to the risk of nephrotoxicity and neuromuscular blockade. If coadministration cannot be avoided, monitor renal and neuromuscular function. Risk D: Consider therapy modification

Cyclizine: May enhance the ototoxic effect of Aminoglycosides. Risk C: Monitor therapy

CycloSPORINE (Systemic): Aminoglycosides may enhance the nephrotoxic effect of CycloSPORINE (Systemic). Risk C: Monitor therapy

Distigmine: Aminoglycosides may diminish the therapeutic effect of Distigmine. Risk C: Monitor therapy

Fecal Microbiota (Live) (Oral): May diminish the therapeutic effect of Antibiotics. Risk X: Avoid combination

Fecal Microbiota (Live) (Rectal): Antibiotics may diminish the therapeutic effect of Fecal Microbiota (Live) (Rectal). Risk X: Avoid combination

Foscarnet: May enhance the nephrotoxic effect of Aminoglycosides. Risk X: Avoid combination

Immune Checkpoint Inhibitors (Anti-PD-1, -PD-L1, and -CTLA4 Therapies): Antibiotics may diminish the therapeutic effect of Immune Checkpoint Inhibitors (Anti-PD-1, -PD-L1, and -CTLA4 Therapies). Risk C: Monitor therapy

Lactobacillus and Estriol: Antibiotics may diminish the therapeutic effect of Lactobacillus and Estriol. Risk C: Monitor therapy

Loop Diuretics: May enhance the adverse/toxic effect of Aminoglycosides. Specifically, nephrotoxicity and ototoxicity. Risk C: Monitor therapy

Mannitol (Systemic): May enhance the nephrotoxic effect of Aminoglycosides. Risk X: Avoid combination

Mecamylamine: Aminoglycosides may enhance the neuromuscular-blocking effect of Mecamylamine. Risk X: Avoid combination

Methoxyflurane: Aminoglycosides may enhance the nephrotoxic effect of Methoxyflurane. Risk X: Avoid combination

Netilmicin (Ophthalmic): Aminoglycosides may enhance the nephrotoxic effect of Netilmicin (Ophthalmic). Risk X: Avoid combination

Neuromuscular-Blocking Agents: Aminoglycosides may enhance the therapeutic effect of Neuromuscular-Blocking Agents. Risk C: Monitor therapy

Nonsteroidal Anti-Inflammatory Agents: May decrease the excretion of Aminoglycosides. Data only in premature infants. Risk C: Monitor therapy

Oxatomide: May enhance the ototoxic effect of Aminoglycosides. Risk C: Monitor therapy

Penicillins: May decrease the serum concentration of Aminoglycosides. Primarily associated with extended spectrum penicillins, and patients with renal dysfunction. Risk C: Monitor therapy

Polymyxin B: May enhance the nephrotoxic effect of Aminoglycosides. Polymyxin B may enhance the neurotoxic effect of Aminoglycosides. Risk X: Avoid combination

Sodium Picosulfate: Antibiotics may diminish the therapeutic effect of Sodium Picosulfate. Management: Consider using an alternative product for bowel cleansing prior to a colonoscopy in patients who have recently used or are concurrently using an antibiotic. Risk D: Consider therapy modification

Tacrolimus (Systemic): Aminoglycosides may enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Tenofovir Products: Aminoglycosides may increase the serum concentration of Tenofovir Products. Tenofovir Products may increase the serum concentration of Aminoglycosides. Risk C: Monitor therapy

Typhoid Vaccine: Antibiotics may diminish the therapeutic effect of Typhoid Vaccine. Only the live attenuated Ty21a strain is affected. Management: Avoid use of live attenuated typhoid vaccine (Ty21a) in patients being treated with systemic antibacterial agents. Postpone vaccination until 3 days after cessation of antibiotics and avoid starting antibiotics within 3 days of last vaccine dose. Risk D: Consider therapy modification

Vancomycin: May enhance the nephrotoxic effect of Aminoglycosides. Vancomycin may enhance the neurotoxic effect of Aminoglycosides. Management: Consider avoiding coadministration of aminoglycosides and vancomycin unless clinically indicated. If coadministered, monitor closely for signs of nephrotoxicity and neurotoxicity. Risk D: Consider therapy modification

Pregnancy Considerations

Gentamicin crosses the placenta.

Aminoglycosides may cause fetal harm if administered to a pregnant patient. There are several reports of total irreversible bilateral congenital deafness in children whose mothers received another aminoglycoside (streptomycin) during pregnancy. Although serious side effects to the fetus/infant have not been reported following maternal use of all aminoglycosides, a potential for harm exists.

Due to pregnancy-induced physiologic changes, some pharmacokinetic parameters of gentamicin may be altered (Popović 2007).

Gentamicin use has been evaluated for various infections in pregnant patients including the treatment of acute pyelonephritis (Jolley 2010) and as an alternative antibiotic for prophylactic use prior to cesarean delivery (Bratzler 2013).

Gentamicin is used in the management of plague (Yersinia pestis). Untreated infections in pregnant patients may result in hemorrhage (including postpartum hemorrhage), maternal and fetal death, preterm birth, and stillbirth. Limited data suggest maternal-fetal transmission of Y. pestis can occur if not treated. Pregnant patients should be treated for Y. pestis; parenteral antibiotics are preferred for initial treatment when otherwise appropriate. Gentamicin (in combination with a fluoroquinolone) is recommended for treating pregnant patients with bubonic, pharyngeal, pneumonic, or septicemic plague (CDC [Nelson 2021]).

Monitoring Parameters

Urinalysis, urine output, BUN, SCr, hearing test (especially for those at risk for ototoxicity or who will be receiving prolonged therapy [>2 weeks]).

Serum gentamicin concentrations:

Conventional dosing: Initial serum concentrations typically obtained after the third dose; may consider obtaining serum concentrations sooner in neonates or patients with rapidly changing renal function.

Extended-interval dosing: Initial serum concentrations may be obtained following the first or second dose (Begg 1999; Knoderer 2003).

Patients receiving outpatient therapy: Serum gentamicin concentrations (at least weekly); CBC with differential (weekly); basic metabolic panel including potassium, creatinine, and BUN (twice weekly) (IDSA [Norris 2019]).

Not all pediatric patients who receive aminoglycosides require monitoring of serum aminoglycoside concentrations. Indications for use of aminoglycoside serum concentration monitoring include:

Treatment course >5 days.

Patients with decreased or changing renal function.

Patients with poor therapeutic response.

Neonates and Infants <3 months of age.

Atypical body constituency (obesity, expanded extracellular fluid volume).

Clinical need for higher doses or shorter intervals (eg, cystic fibrosis, burns, endocarditis, meningitis, critically ill patients, relatively resistant organism).

Patients on hemodialysis or chronic ambulatory peritoneal dialysis.

Signs of nephrotoxicity or ototoxicity.

Concomitant use of other nephrotoxic agents.

Some beta-lactam agents (penicillins and cephalosporins) may accelerate the degradation of aminoglycosides in vitro; this may be clinically significant in patients with significant renal impairment. Close monitoring of aminoglycoside concentrations and clinical response is warranted.

Reference Range

Note: Sampling times and target concentrations provided reflect general practices; patient-specific factors, including type and site of infection, goals of therapy, severity of illness, clinical status, fluid status, and kidney function should be considered when determining patient-specific therapeutic monitoring plans and targets. Outcomes are predicted by serum concentrations and the minimum inhibitory concentration (MIC) of the infecting organism. Efficacy has been associated with peak:MIC ratios of ≥8 to 10 and AUC24:MIC ratios of 30 to 100 depending on severity of infection (Bland 2018; Craig 2011; Knoderer 2003). Nephrotoxicity is associated with elevated trough concentrations (Bertino 1993; Rybak 1999). Some recommendations are based on tobramycin:

Neonates:

Timing of serum samples (El-Chaar 2016; Touw 2009; van Donge 2018; van Maarseveen 2016; manufacturer's labeling):

Peak: 30 minutes after end of 30-minute infusion, or 1 hour following start of infusion or IM injection.

Trough: End of dosing interval, just prior to the next dose.

Target concentrations: Note: Studies evaluating extended-interval dosing in neonates typically target concentrations more often associated with conventional dosing (Miller 2014).

Peak: ≥8 to 12 mg/L; 10 x MIC (El-Chaar 2016; Touw 2009; van Maarseveen 2016).

Trough: <0.5 to 1 mg/L (Touw 2009; van Maarseveen 2016).

Infants, Children, and Adolescents:

Conventional dosing:

Timing of serum samples:

Peak: 30 minutes after 30-minute infusion, or 1 hour following start of infusion or IM injection.

Trough: End of dosing interval, immediately before next dose.

Target concentrations:

Peak:

General: 6 to 12 mg/L or 10 x MIC (Red Book [AAP 2021]). Note: Peak concentrations on the lower end of the range would only achieve efficacy target (10 x MIC) for MICs ≤0.5 mg/L.

Urinary tract infections: 4 to 6 mg/L (Matzke 1983).

Synergy against gram-positive organisms: 3 to 4 mg/L (AHA [Baltimore 2015]).

Trough:

General: <1 to 2 mg/L (Hammett-Stabler 1998; Red Book [AAP 2021]); others suggest a target of <0.5 to 1 mg/L (Bland 2018; Touw 2009).

Synergy against gram-positive organisms: <1 mg/L (AHA [Baltimore 2015]).

Extended-interval dosing: Note: Monitoring protocols are not standardized; various approaches exist. In addition to the strategies discussed, AUC monitoring has also been described (Begg 1999; Bland 2018; Roy 2016).

Timing of serum samples:

Peak: 30 minutes after the end of a 30-minute infusion (Jenh 2011; Knoderer 2003). Some suggest waiting ≥60 minutes after the end of the infusion to allow time for distribution of higher doses and using patient-specific pharmacokinetic parameters to back-extrapolate true peak concentration (Prescott 2010; Safi 2016). It is not always necessary to monitor peak concentration depending on clinical situation (Begg 1999; Jenh 2011).

Trough: Obtain at the end of a dosing interval, just prior to the next dose (Begg 1999; Knoderer 2003). May also obtain 4 hours prior to the next dose to ensure that drug has cleared (McDade 2010).

Patient-specific calculations: Two concentrations at least 1 half-life apart can be obtained (eg, a peak and a mid-interval concentration 8 to 12 hours after the dose, or 2 postdistribution concentrations while drug is still detectable) to calculate patient-specific pharmacokinetic parameters and extrapolate peak, trough, AUC, and/or drug-free interval (Jenh 2011; Knoderer 2003). For patients with cystic fibrosis, the 2 random samples postinfusion method is the most common strategy used to monitor aminoglycosides (eg, obtain a serum concentration 2 hours and 8 to 10 hours after the start of the infusion, calculate elimination rate, and extrapolate peak and trough) (Zobell 2016).

Target concentrations:

Patients without cystic fibrosis:

Peak: 8 to 10 x MIC of the pathogen (~15 to 35 mg/L) (Jenh 2011; Knoderer 2003; Roy 2016).

Trough: <0.5 to 1 mg/L; ideally undetectable; a drug-free period is desired (Jenh 2011; Knoderer 2003; McDade 2010).

Patients with cystic fibrosis:

Peak: 20 to 30 mg/L (Safi 2016; Smyth 2005); target range variable; higher target concentrations up to 40 mg/L have been reported (Flume 2009; Zobell 2016). May be necessary to modify target based on MIC to achieve targets associated with efficacy (Burkhardt 2006).

Trough: <0.5 to 1 mg/L; undetectable (Flume 2009; Safi 2016; Wassil 2008; Zobell 2016). A drug-free period is desired; some centers adjust dose to provide a drug-free interval of ≤8 to 12 hours (Zobell 2016).

AUC: 75 to 125 mg•hour/L; most centers target 80 to 120 mg•hour/L (Zobell 2016).

Mechanism of Action

Interferes with bacterial protein synthesis by binding to 30S ribosomal subunit resulting in a defective bacterial cell membrane

Pharmacokinetics (Adult Data Unless Noted)

Absorption: IM: Rapid and complete; Oral: Poorly absorbed (<1%) (MacDougall 2011).

Distribution: Primarily into extracellular fluid (highly hydrophilic); high concentration in the renal cortex; minimal penetration to CSF and ocular tissues via IV route.

Vd: Varies with age; increased by edema, ascites, fluid overload; decreased with dehydration:

Neonates: 0.45 ± 0.1 L/kg.

Infants: 0.4 ± 0.1 L/kg.

Children: 0.35 ± 0.15 L/kg.

Adolescents: 0.3 ± 0.1 L/kg.

Adults: 0.2 to 0.3 L/kg (Leggett 2015; Xuan 2004).

CSF:blood level ratio: Normal meninges: <10%; Inflamed meninges: ≤25% (MacDougall 2011)

Lung:

Bronchial secretion Cmax (peak): serum Cmax (peak) ratio: ~50%, varies with time and route (IM vs IV) (Pennington 1975).

Epithelial lining fluid Cmax (peak): serum Cmax (peak) ratio: ~32%, varies with time (Heffernan 2019; Panidis 2005; Rodvold 2011).

Protein binding: <30%.

Half-life elimination:

Neonates: <1 week: 3 to 11.5 hours; 1 week to 1 month: 3 to 6 hours.

Infants: 4 ± 1 hour.

Children: 2 ± 1 hour.

Adolescents: 1.5 ± 1 hour.

Adults: ~2 hours (Regamey 1973); Renal failure: mean: 41 ± 24 hours; Range: 6 to 127 hours (Dager 2006).

Time to peak, serum: IM: 30 to 90 minutes; IV: 30 minutes after 30-minute infusion (MacDougall 2011); Note: Distribution is prolonged after larger doses (≥90 minutes after 30- to 60-minute infusion of 7 mg/kg) (Demczar 1997; McNamara 2001; Wallace 2002).

Excretion: Urine (≥70% as unchanged drug).

Clearance: Directly related to renal function.

Neonates: 0.045 ± 0.01 L/hour/kg.

Infants: 0.1 ± 0.05 L/hour/kg.

Children: 0.1 ± 0.03 L/hour/kg.

Adolescents: 0.09 ± 0.03 L/hour/kg.

Adults: 0.06 L/hour/kg (Xuan 2004).

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Altered kidney function: Clearance is decreased in renal impairment.

Anti-infective considerations:

Parameters associated with efficacy: Gram-negative bacilli:Concentration-dependent; associated with Cmax (peak)/minimum inhibitory concentration (MIC), goal: ≥8 to 10 (Kashuba 1999; Moore 1987; Zelenitsky 2003) or AUC24/MIC, goal: 30 to 50 (mild/moderate infection, immunocompetent patient) or 80 to 100 (severe infection) (Bland 2018; Craig 2011; Drusano 2007; Nielsen 2011).

Expected drug exposure in patients with normal renal function:

Cmax (peak), postdistributional:

Infants, children, and adolescents (varies with age; values are generalized):

Neutropenic fever: 7 mg/kg: ~17 mg/L (Shankar 1999).

Critically ill: 7 to 8 mg/kg: 15 to 22 mg/L (Lopez 2010).

1.5 to 2.5 mg/kg: ~5 to 6 mg/L (Alsultan 2019; Knoderer 2003).

Adults:

7 mg/kg: ~22 mg/L (Finnell 1998; Xuan 2004).

2 mg/kg: ~6 mg/L (Demczar 1997).

AUC24:

Children and adolescents:

Neutropenic fever: 7 to 8 mg/kg: ~45 to 80 mg•hour/L (Bialkowski 2016).

Adults:

7 mg/kg: ~70 to 110 mg•hour/L (Barclay 1995; Craig 2011; Finnell 1998).

Parameters associated with toxicity: Nephrotoxicity is associated with more frequent administration and elevated Cmin (trough) concentrations leading to renal accumulation (Bertino 1993; Rybak 1999; Verpooten 1989); AUC has also been found to be predictive (Rybak 1999).

Postantibiotic effect: Bacterial killing continues after gentamicin concentration drops below the MIC of targeted pathogen; generally ~0.5 to 12 hours, though the actual time of postantibiotic effect varies significantly based on multiple factors including organism, gentamicin Cmax (peak), and concomitant antimicrobial therapy (Craig 2011; Gudmundsson 1993; Lacy 1998; Urban 1997).

Brand Names: International
International Brand Names by Country
For country code abbreviations (show table)

  • (AE) United Arab Emirates: Garamycin;
  • (AR) Argentina: Gentamicina | Gentamicina Larjan | Gentamicina richet | Gentamicina Rivero | Gentamina | Glevomicina | Plurisemina;
  • (AT) Austria: Gentamicin B Braun;
  • (AU) Australia: Gentamicin | Gentamicin sulphate | Gentamycin;
  • (BD) Bangladesh: Garamycin | Gentamycin | Genticin | Gentin | Invigen;
  • (BE) Belgium: Gentamycine b.braun | Gentamycine baxter | Geomycine;
  • (BR) Brazil: Garacin;
  • (CL) Chile: Gentalyn | Gentamicina;
  • (CO) Colombia: Garamicina | Gentabiotic | Gentamicina;
  • (CZ) Czech Republic: Gentamicin braun | Gentamycin k;
  • (DE) Germany: Gentamicin B Braun;
  • (DO) Dominican Republic: Garamicina | Gentamicina | Servigenta;
  • (EC) Ecuador: Gentabrand | Gentamicina | Gentastar;
  • (EE) Estonia: Gentamicin;
  • (EG) Egypt: Garamycin | Regaminol;
  • (ES) Spain: Genta Gobens | Gentamicina Braun | Gentamicina grifol | Gentamicina normon | Genticina | Gevramycin;
  • (FI) Finland: Garamycin | Gentamicin B Braun;
  • (FR) France: Gentamicine panpharma;
  • (GB) United Kingdom: Gentamicin | Trav gentamicin;
  • (GR) Greece: Gentamedin;
  • (HK) Hong Kong: Gentamicin;
  • (ID) Indonesia: Gentamicin sulfat;
  • (IE) Ireland: Cidomycin | Genticin;
  • (IN) India: Gentamycin;
  • (IT) Italy: Gentalyn | Gentamen | Gentamicina b. braun;
  • (JO) Jordan: Cidomycin | Garamycin;
  • (JP) Japan: Geminimycin | Gentacin | Gentamic.sulf.show;
  • (KR) Korea, Republic of: Gentamicin;
  • (KW) Kuwait: Garamycin;
  • (LB) Lebanon: Garamycin;
  • (LT) Lithuania: Gentamycin k;
  • (LU) Luxembourg: Gentamicin;
  • (LV) Latvia: Gentamycin | Gentamycin k;
  • (MA) Morocco: Gentalline | Gentamen | Gentamycine llorente | Gentosyl | Polybac;
  • (MX) Mexico: Barmicil | Fustermicina | Garalen | Gentacin | Gentamicina delta | Tondex;
  • (MY) Malaysia: Garamycin | Gentamicin | Gentamycin;
  • (NL) Netherlands: Gentamicinum;
  • (NO) Norway: Gensumycin | Gentamicin | Gentamicin braun | Gentamicina;
  • (NZ) New Zealand: Gentamicin;
  • (PE) Peru: Azupel;
  • (PH) Philippines: Adelanin | Gentaxin | Obogen | Rocygen;
  • (PK) Pakistan: Genacyn | Gentamicin | Gentamycin | Refobacin;
  • (PL) Poland: Gentamicin b.braun | Gentamycin;
  • (PR) Puerto Rico: Garamycin | Gentamicin;
  • (PT) Portugal: Garalone | Gentamicina | Gentamicina b. braun;
  • (PY) Paraguay: Gentamedin;
  • (QA) Qatar: Gentam | Gentamed | Genticyn | Gentthaver;
  • (RO) Romania: Gentamycin k;
  • (RU) Russian Federation: Gentamicin ferein | Gentamicin k;
  • (SA) Saudi Arabia: Cidomycin | Genta;
  • (SG) Singapore: Gentamicin | Gentamicin sulphate | Gentamycin | Genticin | Miramycin;
  • (SI) Slovenia: Gentamicin B Braun;
  • (SK) Slovakia: Gentamicim B.braun | Gentamycin k;
  • (TH) Thailand: Cidomycin | Garamycin | Gentacin | Gentamicin | Gentamicin injection meiji | Gentamycin | Gentawin | Getamic;
  • (TN) Tunisia: Gentamicine | Gentosyl;
  • (TR) Turkey: Getamisin;
  • (TW) Taiwan: Cantamycin | Garamycin | Genta Gobens | Gentamicin | Gentamycin | Gentamycin sulfate | Gentapromes | Larkmycin | Miramycin | Patamicin | Servigenta;
  • (UA) Ukraine: Gentamycin k;
  • (UY) Uruguay: Gentamina | Migenta;
  • (VE) Venezuela, Bolivarian Republic of: Catogen | Gentalyn inyectabl | Gentamicina | Gentisul;
  • (ZA) South Africa: Gentamycin | Gentamycin-fresenius | Sabax gentamix
  1. Alsultan A, Abouelkheir M, Elsharawy Y, et al. Optimizing gentamicin dosing in pediatrics using Monte Carlo simulations. Pediatr Infect Dis J. 2019;38(4):390-395. doi:10.1097/INF.0000000000002120 [PubMed 30882729]
  2. American Academy of Pediatrics (AAP). In: Kimberlin DW, Barnett ED, Lynfield R, Sawyer MH, eds. Red Book: 2021-2024 Report of the Committee on Infectious Diseases. 32nd ed. American Academy of Pediatrics; 2021.
  3. American Academy of Pediatrics, Subcommittee on Urinary Tract Infection, Steering Committee on Quality Improvement and Management, Roberts KB. Urinary tract infection: clinical practice guideline for the diagnosis and management of the initial UTI in febrile infants and children 2 to 24 months. Pediatrics. 2011;128(3):595-610. [PubMed 21873693]
  4. American College of Obstetricians and Gynecologists (ACOG). Committee on Obstetric Practice. Committee opinion No. 712: intrapartum management of intraamniotic infection. Obstet Gynecol. 2017;130(2):e95-e101. doi:10.1097/AOG.0000000000002236 [PubMed 28742677]
  5. American College of Obstetricians and Gynecologists (ACOG). Committee on Practice Bulletins-Obstetrics. ACOG practice bulletin No. 199: use of prophylactic antibiotics in labor and delivery. Obstet Gynecol. 2018;132(3):e103-e119. doi:10.1097/AOG.0000000000002833 [PubMed 30134425]
  6. Anderson DJ, Sexton DJ. Antimicrobial prophylaxis for prevention of surgical site infections in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  7. Anderson PO, Sauberan JB. Modeling drug passage into human milk. Clin Pharmacol Ther. 2016;100(1):42-52. [PubMed 27060684]
  8. Ariza J, Bosilkovski M, Cascio A, et al; International Society of Chemotherapy; Institute of Continuing Medical Education of Ioannina. Perspectives for the treatment of brucellosis in the 21st century: the Ioannina recommendations. PLoS Med. 2007;12(4):1872-1878. [PubMed 18162038]
  9. Aronoff GR, Bennett WM, Berns JS, et al. Drug Prescribing in Renal Failure: Dosing Guidelines for Adults and Children. 5th ed. American College of Physicians, 2007.
  10. Autore G, Bernardi L, La Scola C, et al. Management of pediatric urinary tract infections: a Delphi study. Antibiotics (Basel). 2022;11(8):1122. doi:10.3390/antibiotics11081122 [PubMed 36009990]
  11. Baddour LM, Flynn PM, Fekete T. Infections of cerebrospinal fluid shunts and other devices. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed April 17, 2019.
  12. Baddour LM, Wilson WR, Bayer AS, et al; American Heart Association Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young; Council on Clinical Cardiology; Council on Cardiovascular Surgery and Anesthesia; and Stroke Council. Infective endocarditis in adults: diagnosis, antimicrobial therapy, and management of complications: a scientific statement for healthcare professionals from the American Heart Association [published corrections appear in Circulation. 2015;132(17):e215; Circulation. 2016;134(8):e113; Circulation. 201831;138(5):e78-e79]. Circulation. 2015;132(15):1435-1486. [PubMed 26373316]
  13. Bailey TC, Little JR, Littenberg B, Reichley RM, Dunagan WC. A meta-analysis of extended-interval dosing versus multiple daily dosing of aminoglycosides. Clin Infect Dis. 1997;24(5):786-795. [PubMed 9142771]
  14. Balighian E, Burke M. Urinary tract infections in children. Pediatr Rev. 2018;39(1):3-12. [PubMed 29292282]
  15. Baltimore RS, Gewitz M, Baddour LM, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young and the Council on Cardiovascular and Stroke Nursing. Infective endocarditis in childhood: 2015 update: a scientific statement from the American Heart Association. Circulation. 2015;132(15):1487-1515. doi:10.1161/CIR.0000000000000298 [PubMed 26373317]
  16. Barclay ML, Duffull SB, Begg EJ, Buttimore RC. Experience of once-daily aminoglycoside dosing using a target area under the concentration-time curve. Aust N Z J Med. 1995;25(3):230-235. doi:10.1111/j.1445-5994.1995.tb01529.x [PubMed 7487691]
  17. Battistella M, Vercaigne LM, Cote D, Lok CE. Antibiotic lock: in vitro stability of gentamicin and sodium citrate stored in dialysis catheters at 37 degrees C. Hemodial Int. 2010;14(3):322-326. doi:10.1111/j.1542-4758.2010.00440.x [PubMed 20345391]
  18. Begg EJ, Barclay ML, Kirkpatrick CJ. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharmacol. 1999;47(1):23-30. [PubMed 10073735]
  19. Bergenwall M, Walker SAN, Elligsen M, et al. Optimizing gentamicin conventional and extended interval dosing in neonates using Monte Carlo simulation - a retrospective study. BMC Pediatr. 2019;19(1):318. [PubMed 31492162]
  20. Berríos-Torres SI, Umscheid CA, Bratzler DW, et al; Healthcare Infection Control Practices Advisory Committee. Centers for Disease Control and Prevention guideline for the prevention of surgical site infection, 2017. JAMA Surg. 2017;152(8):784-791. doi: 10.1001/jamasurg.2017.0904. [PubMed 28467526]
  21. Bertino JS Jr, Booker LA, Franck PA, Jenkins PL, Franck KR, Nafziger AN. Incidence of and significant risk factors for aminoglycoside-associated nephrotoxicity in patients dosed by using individualized pharmacokinetic monitoring. J Infect Dis. 1993;167(1):173-179. doi:10.1093/infdis/167.1.173 [PubMed 8418164]
  22. Bertino JS Jr, Rodvold KA, Destache CJ. Cost considerations in therapeutic drug monitoring of aminoglycosides. Clin Pharmacokinet. 1994;26(1):71-81. [PubMed 8137600]
  23. Bhatt-Mehta V, Johnson CE and Schumacher RE, “Gentamicin Pharmacokinetics in Term Neonates Receiving Extracorporeal Membrane Oxygenation,” Pharmacotherapy, 1992, 12(1):28-32. [PubMed 1549536]
  24. Bialkowski S, Staatz CE, Clark J, Lawson R, Hennig S. Gentamicin pharmacokinetics and monitoring in pediatric patients with febrile neutropenia. Ther Drug Monit. 2016;38(6):693-698. doi:10.1097/FTD.0000000000000341 [PubMed 27851686]
  25. Bilbao-Meseguer I, Rodríguez-Gascón A, Barrasa H, Isla A, Solinís MÁ. Augmented renal clearance in critically ill patients: a systematic review. Clin Pharmacokinet. 2018;57(9):1107-1121. doi:10.1007/s40262-018-0636-7 [PubMed 29441476]
  26. Blackburn LM, Tverdek FP, Hernandez M, Bruno JJ. First-dose pharmacokinetics of aminoglycosides in critically ill haematological malignancy patients. Int J Antimicrob Agents. 2015;45(1):46-53. doi: 10.1016/j.ijantimicag.2014.09.006. [PubMed 25455848]
  27. Bland CM, Pai MP, Lodise TP. Reappraisal of contemporary pharmacokinetic and pharmacodynamic principles for informing aminoglycoside dosing. Pharmacotherapy. 2018;38(12):1229-1238. doi:10.1002/phar.2193 [PubMed 30403305]
  28. Bosilkovski M. Brucellosis: treatment, and prevention. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 31, 2022.
  29. Boulanger LL, Ettestad P, Fogarty JD, Dennis DT, Romig D, Mertz G. Gentamicin and tetracyclines for the treatment of human plague: review of 75 cases in New Mexico, 1985-1999. Clin Infect Dis. 2004;38(5):663-669. [PubMed 14986250]
  30. Bradley JS, Nelson JD, Barnett ED, et al, eds. Nelson's Pediatric Microbial Therapy. 27th ed. American Academy of Pediatrics; 2021.
  31. Bratzler DW, Dellinger EP, Olsen KM, et al; American Society of Health-System Pharmacists; Infectious Diseases Society of America; Surgical Infection Society; Society for Healthcare Epidemiology of America. Clinical practice guidelines for antimicrobial prophylaxis in surgery. Am J Health Syst Pharm. 2013;70(3):195-283. [PubMed 23327981]
  32. Bremmer D, Bookstaver B, Cairns M, et al. Impact of body mass index and bacterial resistance in osteomyelitis after antibiotic prophylaxis of open lower-extremity fractures. Surg Infect (Larchmt). 2017;18(3):368-373. doi:10.1089/sur.2016.219 [PubMed 28394750]
  33. Buck ML, "Pharmacokinetic Changes During Extracorporeal Membrane Oxygenation: Implications for Drug Therapy of Neonates," Clin Pharmacokinet, 2003, 42(5):403-17. [PubMed 12739981]
  34. Buijk SE, Mouton JW, Gyssens IC, Verbrugh HA, Bruining HA. Experience with a once-daily dosing program of aminoglycosides in critically ill patients. Intensive Care Med. 2002;28(7):936-942. [PubMed 12122533]
  35. Burkart JM. Microbiology and therapy of peritonitis in peritoneal dialysis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 21, 2022.
  36. Burkhardt O, Lehmann C, Madabushi R, Kumar V, Derendorf H, Welte T. Once-daily tobramycin in cystic fibrosis: better for clinical outcome than thrice-daily tobramycin but more resistance development? J Antimicrob Chemother. 2006;58(4):822-829. doi:10.1093/jac/dkl328 [PubMed 16885180]
  37. Bury RG and Tudehope D, "Enteral Antibiotics for Preventing Necrotizing Enterocolitis in Low Birthweight or Preterm Infants," Cochrane Database Syst Rev, 2001, (1):CD000405. [PubMed 11279690]
  38. Carapetis JR, Jaquiery AL, Buttery JP, et al, "Randomized, Controlled Trial Comparing Once Daily and Three Times Daily Gentamicin in Children With Urinary Tract Infections," Pediatr Infect Dis J, 2001, 20(3):240-6. [PubMed 11303823]
  39. Celiloglu M, Celiker S, Guven H, et al, "Gentamicin Excretion and Uptake From Breast Milk by Nursing Infants," Obstet Gynecol, 1994, 84(2):263-5. [PubMed 8041544]
  40. Centers for Disease Control and Prevention (CDC). Recommended antibiotic treatment for plague. http://www.cdc.gov/plague/resources/Recommended-antibiotics-for-plague_revision-Aug-2015_Final-(00000002).pdf. Updated October 2015.
  41. Chen KT. Postpartum endometritis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 10, 2024.
  42. Chu VH. Antimicrobial therapy of left-sided native valve endocarditis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed December 8, 2020.
  43. Chung AM, Reed MD, Blumer JL. Antibiotics and breast-feeding: a critical review of the literature. Paediatr Drugs. 2002;4(12):817-837. [PubMed 12431134]
  44. Contopoulos-Ioannidis DG, Giotis ND, Baliatsa DV, et al. Extended-interval aminoglycoside administration for children: a meta-Analysis. Pediatrics. 2004;114(1):e111-e118. [PubMed 15231982]
  45. Cook AM, Mieure KD, Owen RD, Pesaturo AB, Hatton J. Intracerebroventricular administration of drugs. Pharmacotherapy. 2009;29(7):832-845. doi: 10.1592/phco.29.7.832 [PubMed 19558257]
  46. Corrections. MMWR Morb Mortal Wkly Rep. 2023;72(4):107-108. doi:10.15585/mmwr.mm7204a5 [PubMed 36701253]
  47. Craig WA. Optimizing aminoglycoside use. Crit Care Clin. 2011;27(1):107-121. doi:10.1016/j.ccc.2010.11.006 [PubMed 21144989]
  48. Dager WE, King JH. Aminoglycosides in intermittent hemodialysis: pharmacokinetics with individual dosing. Ann Pharmacother. 2006;40:9-14. [PubMed 16332944]
  49. Daly JS, Dodge RA, Glew RH, et al, "Effect of Time and Temperature on Inactivation of Aminoglycosides by Ampicillin at Neonatal Dosages," J Perinatol, 1997, 17(1):42-5. [PubMed 9069064]
  50. de Hoog M, Mouton JW, Schoemaker RC, et al, "Extended-Interval Dosing of Tobramycin in Neonates: Implications for Therapeutic Drug Monitoring," Clin Pharmacol Ther, 2002, 71(5):349-58. [PubMed 12011820]
  51. Del Priore G, Jackson-Stone M, Shim EK, Garfinkel J, Eichmann MA, Frederiksen MC. A comparison of once-daily and 8-hour gentamicin dosing in the treatment of postpartum endometritis. Obstet Gynecol. 1996;87(6):994-1000. [PubMed 8649712]
  52. Demczar DJ, Nafziger AN, Bertino JS Jr. Pharmacokinetics of gentamicin at traditional versus high doses: implications for once-daily aminoglycoside dosing. Antimicrob Agents Chemother. 1997;41(5):1115-1119. [PubMed 9145878]
  53. Dennis DT, Inglesby TV, Henderson DA, et al; Working Group on Civilian Biodefense. Tularemia as a biological weapon: medical and public health management. JAMA. 2001;285(21):2763-2773. [PubMed 11386933]
  54. Drew RH. Dosing and administration of parenteral aminoglycosides. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 8, 2023.
  55. Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger AN, Louie A. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect Dis. 2007;45(6):753-760. doi:10.1086/520991 [PubMed 17712761]
  56. El-Chaar GM, Supaswud-Franks T, Venugopalan L, Kohn N, Castro-Alcaraz S. Extended-interval gentamicin administration in neonates: a simplified approach. J Perinatol. 2016;36(8):660-665. [PubMed 26986995]
  57. Evans L, Rhodes A, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Intensive Care Med. 2021;47(11):1181-1247. doi:10.1007/s00134-021-06506-y [PubMed 34599691]
  58. Expert opinion. Senior Renal Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.
  59. Finnell DL, Davis GA, Cropp CD, Ensom MH. Validation of the Hartford nomogram in trauma surgery patients. Ann Pharmacother. 1998;32(4):417-421. doi:10.1345/aph.17243 [PubMed 9562135]
  60. Flume PA, Mogayzel PJ Jr, Robinson KA, et al, "Cystic Fibrosis Pulmonary Guidelines: Treatment of Pulmonary Exacerbations," Am J Respir Crit Care Med, 2009, 180(9):802-8. [PubMed 19729669]
  61. Fortún J, Grill F, Martín-Dávila P, et al. Treatment of long-term intravascular catheter-related bacteraemia with antibiotic-lock therapy. J Antimicrob Chemother. 2006;58(4):816-821. [PubMed 16899468]
  62. Foucault C, Raoult D, Brouqui P. Randomized open trial of gentamicin and doxycycline for eradication of Bartonella quintana from blood in patients with chronic bacteremia. Antimicrob Agents Chemother. 2003;47(7):2204-2207. [PubMed 12821469]
  63. Fox MT, Amoah J, Hsu AJ, Herzke CA, Gerber JS, Tamma PD. Comparative effectiveness of antibiotic treatment duration in children with pyelonephritis. JAMA Netw Open. 2020;3(5):e203951. doi:10.1001/jamanetworkopen.2020.3951 [PubMed 32364593]
  64. Friedman ND, Sexton DJ. Gram-negative bacillary meningitis: Treatment. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  65. Gelfand MS. Treatment, prognosis, and prevention of Listeria monocytogenes infection. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 12, 2020.
  66. Gentamicin in NS Viaflex container [prescribing information]. Deerfield, IL: Baxter Healthcare Corporation; May 2018.
  67. Gentamicin pediatric injection [prescribing information]. Lake Zurich, IL: Fresenius Kabi; October 2019.
  68. Gentamicin MDV injection, USP [prescribing information]. Berkeley Heights, NJ: Hikma Pharmaceuticals USA Inc; February 2022.
  69. Gilbert DN, Lee BL, Dworkin RJ, et al. A randomized comparison of the safety and efficacy of once-daily gentamicin or thrice-daily gentamicin in combination with ticarcillin-clavulanate. Am J Med. 1998;105(3):182-191. [PubMed 9753020]
  70. Girand HL. Antibiotic lock therapy for treatment of catheter-related bloodstream infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  71. Goodlet KJ, Benhalima FZ, Nailor MD. A systematic review of single-dose aminoglycoside therapy for urinary tract infection: is it time to resurrect an old strategy?. Antimicrob Agents Chemother. 2018;63(1):e02165-18. [PubMed 30397061]
  72. Goodwin SD, Nix DE, Heyd A, et al, “Compatibility of Ciprofloxacin Injection with Selected Drugs and Solutions,” Am J Hosp Pharm, 1991, 48(10):2166-71. [PubMed 1781473]
  73. Gould FK, Denning DW, Elliott TS, et al, “Guidelines for the Diagnosis and Antibiotic Treatment of Endocarditis in Adults: A Report of the Working Party of the British Society for Antimicrobial Chemotherapy,” J Antimicrob Chemother, 2012, 67(2):269-89. [PubMed 22086858]
  74. Grylack LJ and Scanlon JW, "Oral Gentamicin Therapy in the Prevention of Neonatal Necrotizing Enterocolitis. A Controlled Double-Blind Trial," Am J Dis Child, 1978, 132(12):1192-4. [PubMed 362900]
  75. Gudmundsson S, Einarsson S, Erlendsdottir H, Moffat J, Bayer W, Craig WA. The post-antibiotic effect of antimicrobial combinations in a neutropenic murine thigh infection model. J Antimicrob Chemother. 1993;31(suppl D):177-191. doi:10.1093/jac/31.suppl_d.177 [PubMed 8335520]
  76. Gupta K. Acute complicated urinary tract infection (including pyelonephritis) in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed March 20, 2021.
  77. Gupta K, Hooton TM, Naber KG, et al; Infectious Diseases Society of America; European Society for Microbiology and Infectious Diseases. International clinical practice guidelines for the treatment of acute uncomplicated cystitis and pyelonephritis in women: a 2010 update by the Infectious Diseases Society of America and the European Society for Microbiology and Infectious Diseases. Clin Infect Dis. 2011;52(5):e103-e120. doi:10.1093/cid/ciq257 [PubMed 21292654]
  78. Hari P, Meena J, Kumar M, et al. Evidence-based clinical practice guideline for management of urinary tract infection and primary vesicoureteric reflux. Pediatr Nephrol. Published online October 28, 2023. doi:10.1007/s00467-023-06173-9 [PubMed 37897526]
  79. Hartman SJF, Orriëns LB, Zwaag SM, Poel T, de Hoop M, de Wildt SN. External validation of model-based dosing guidelines for vancomycin, gentamicin, and tobramycin in critically ill neonates and children: a pragmatic two-center study. Paediatr Drugs. 2020;22(4):433-444. [PubMed 32507958]
  80. Hasanjani Roushan MR, Mohraz M, Hajiahmadi M, Ramzani A, Valayati AA. Efficacy of gentamicin plus doxycycline versus streptomycin plus doxycycline in the treatment of brucellosis in humans. Clin Infect Dis. 2006;42(8):1075-1080. [PubMed 16575723]
  81. Hassan E, Ober JD. Predicted and measured aminoglycoside pharmacokinetic parameters in critically ill patients. Antimicrob Agents Chemother. 1987;31(11):1855-1858. doi:10.1128/aac.31.11.1855 [PubMed 3435131]
  82. Heffernan AJ, Sime FB, Lipman J, et al. Intrapulmonary pharmacokinetics of antibiotics used to treat nosocomial pneumonia caused by gram-negative bacilli: a systematic review. Int J Antimicrob Agents. 2019;53(3):234-245. doi:10.1016/j.ijantimicag.2018.11.011 [PubMed 30472292]
  83. Heintz BH, Matzke GR, Dager WE. Antimicrobial dosing concepts and recommendations for critically ill adult patients receiving continuous renal replacement therapy or intermittent hemodialysis. Pharmacotherapy. 2009;29(5):562-577. doi:10.1592/phco.29.5.562 [PubMed 19397464]
  84. Hilmer SN, Perera V, Mitchell S, et al. The assessment of frailty in older people in acute care. Australas J Ageing. 2009;28(4):182-188. [PubMed 19951339]
  85. Hilmer SN, Tran K, Rubie P, et al. Gentamicin pharmacokinetics in old age and frailty. Br J Clin Pharmacol. 2011;71(2):224-231. [PubMed 21219402]
  86. Hodiamont CJ, Janssen JM, de Jong MD, Mathôt RA, Juffermans NP, van Hest RM. Therapeutic drug monitoring of gentamicin peak concentrations in critically ill patients. Ther Drug Monit. 2017;39(5):522-530. doi: 10.1097/FTD.0000000000000432. [PubMed 28682925]
  87. Hoff WS, Bonadies JA, Cachecho R, Dorlac WC. East Practice Management Guidelines Work Group: update to practice management guidelines for prophylactic antibiotic use in open fractures. J Trauma. 2011;70(3):751-754. doi:10.1097/TA.0b013e31820930e5 [PubMed 21610369]
  88. Ito S. Drug therapy for breast-feeding women. N Engl J Med. 2000;343(2):118-126. [PubMed 10891521]
  89. Jenh AM, Tamma PD, Milstone AM. Extended-interval aminoglycoside dosing in pediatrics. Pediatr Infect Dis J. 2011;30(4):338-339. [PubMed 21407038]
  90. Jolley JA, Wing DA. Pyelonephritis in pregnancy: an update on treatment options for optimal outcomes. Drugs. 2010;70(13):1643-1655. [PubMed 20731473]
  91. Kaiser AB, McGee ZA. Aminoglycoside therapy of gram-negative bacillary meningitis. N Engl J Med. 1975;293(24):1215-1220. doi:10.1056/NEJM197512112932401 [PubMed 1102982]
  92. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society [published online July 14, 2016]. Clin Infect Dis. doi: 10.1093/cid/ciw353. [PubMed 27418577]
  93. Kanj SS, Sexton DJ. Principles of antimicrobial therapy of Pseudomonas aeruginosa infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 18, 2020.
  94. Karchmer AW, Chu VH. Antimicrobial therapy of prosthetic valve endocarditis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2020.
  95. Kashuba AD, Nafziger AN, Drusano GL, Bertino JS Jr. Optimizing aminoglycoside therapy for nosocomial pneumonia caused by gram-negative bacteria. Antimicrob Agents Chemother. 1999;43(3):623-629. [PubMed 10049277]
  96. Khan AJ, Kumar K, Evans HE. Single-dose gentamicin therapy of recurrent urinary tract infection in patients with normal urinary tracts. J Pediatr. 1987;110(1):131-135. [PubMed 3794874]
  97. Kirkcaldy RD, Weinstock HS, Moore PC, et al. The efficacy and safety of gentamicin plus azithromycin and gemifloxacin plus azithromycin as treatment of uncomplicated gonorrhea. Clin Infect Dis. 2014;59(8):1083-1091. doi:10.1093/cid/ciu521 [PubMed 25031289]
  98. Knoderer CA, Everett JA, Buss WF. Clinical issues surrounding once-daily aminoglycoside dosing in children. Pharmacotherapy. 2003;23(1):44-56. doi:10.1592/phco.23.1.44.31924 [PubMed 12523459]
  99. König K, Lim A, Miller A, Saker S, Guy KJ, Barfield CP. Gentamicin trough levels using a simplified extended-interval dosing regimen in preterm and term newborns. Eur J Pediatr. 2015;174(5):669-673. doi:10.1007/s00431-014-2450-z [PubMed 25388408]
  100. Koshida R, Nakashima E, Taniguchi N, Tsuji A, Benet LZ, Ichimura F. Prediction of the distribution volumes of cefazolin and tobramycin in obese children based on physiological pharmacokinetic concepts. Pharm Res. 1989;6(6):486-491. [PubMed 2762224]
  101. Kraus DM, Pai MP, and Rodvold KA, "Efficacy and Tolerability of Extended-Interval Aminoglycoside Administration in Pediatric Patients," Paediatr Drugs, 2002, 4(7):469-84. [PubMed 12083974]
  102. Krishnasami Z, Carlton D, Bimbo L, et al. Management of hemodialysis catheter-related bacteremia with an adjunctive antibiotic lock solution. Kidney Int. 2002;61(3):1136-1142. [PubMed 11849468]
  103. Lacy MK, Nicolau DP, Nightingale CH, Quintiliani R. The pharmacodynamics of aminoglycosides. Clin Infect Dis. 1998;27(1):23-27. doi:10.1086/514620 [PubMed 9675444]
  104. Leggett JE. Aminoglycosides. In: Bennett JE, Doin R, Blaser MJ, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Philadelphia, PA: Saunders; 2015.
  105. Leroy A, Humbert G, Oksenhendler G, Fillastre JP. Pharmacokinetics of aminoglycosides in subjects with normal and impaired renal function. Antibiot Chemother (1971). 1978;25:163-180. doi:10.1159/000401061 [PubMed 352252]
  106. Li PK, Szeto CC, Piraino B, et al; International Society for Peritoneal Dialysis. Peritoneal dialysis-related infections recommendations: 2010 update [published correction appears in Perit Dial Int. 2011;31(5):512]. Perit Dial Int. 2010;30(4):393-423. doi: 10.3747/pdi.2010.00049. [PubMed 20628102]
  107. Li PK, Szeto CC, Piraino B, et al. ISPD peritonitis recommendations: 2016 update on prevention and treatment. Perit Dial Int. 2016;36(5):481-508. [PubMed 27282851]
  108. Livingston JC, Llata E, Rinehart E, et al. Gentamicin and clindamycin therapy in postpartum endometritis: the efficacy of daily dosing versus dosing every 8 hours. Am J Obstet Gynecol. 2003;188(1):149-152. [PubMed 12548209]
  109. Locksmith GJ, Chin A, Vu T, Shattuck KE, Hankins GD. High compared with standard gentamicin dosing for chorioamnionitis: a comparison of maternal and fetal serum drug levels. Obstet Gynecol. 2005;105(3):473-479. [PubMed 15738010]
  110. Lopez SA, Mulla H, Durward A, Tibby SM. Extended-interval gentamicin: population pharmacokinetics in pediatric critical illness. Pediatr Crit Care Med. 2010;11(2):267-274. doi:10.1097/PCC.0b013e3181b80693 [PubMed 19770786]
  111. MacDougall C, Chambers HF. Aminoglycosides. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics, 12th ed, Chapter 54, Brunton LL, Chabner BA, Knollmann BC, eds. McGraw-Hill, 2011.
  112. Machuca I, Gutiérrez-Gutiérrez B, Gracia-Ahufinger I, et al. Mortality associated with bacteremia due to colistin-resistant Klebsiella pneumoniae with high-level meropenem resistance: importance of combination therapy without colistin and carbapenems. Antimicrob Agents Chemother. 2017;61(8). pii: e00406-17. doi: 10.1128/AAC.00406-17. [PubMed 28559247]
  113. Mackeen AD, Packard RE, Ota E, Speer L. Antibiotic regimens for postpartum endometritis. Cochrane Database Syst Rev. 2015;(2):CD001067. doi: 10.1002/14651858.CD001067.pub3. [PubMed 25922861]
  114. Mancini A, Todd L. Inconsistencies in ISPD peritonitis recommendations: 2016 update on prevention and treatment and the ISPD catheter-related infection recommendations: 2017 update. Perit Dial Int. 2018;38(4):309-310. doi: 10.3747/pdi.2018.00026. [PubMed 29987068]
  115. Mangi RJ, Holstein LL, Andriole VT. Treatment of gram-negative bacillary meningitis with intrathecal gentamicin. Yale J Biol Med. 1977;50(1):31-41. [PubMed 322399]
  116. Mann HJ, Fuhs DW, Awang R, et al, “Altered Aminoglycoside Pharmacokinetics in Critically Ill Patients With Sepsis,” Clin Pharm, 1987, 6(2):148-53. [PubMed 3665367]
  117. Martingano D, Renson A, Rogoff S, et al. Daily gentamicin using ideal body weight demonstrates lower risk of postpartum endometritis and increased chance of successful outcome compared with traditional 8-hour dosing for the treatment of intrapartum chorioamnionitis. J Matern Fetal Neonatal Med. 2019;32(19):3204-3208. doi: 10.1080/14767058.2018.1460348. [PubMed 29642754]
  118. Matz GJ, “Aminoglycoside Ototoxicity,” Am J Otolaryngol, 1986, 7(2):117-9. [PubMed 3485932]
  119. Matzke GR, Burkle WS, Lucarotti RL. Gentamicin and tobramycin dosing guidelines: an evaluation. Drug Intell Clin Pharm.1983;17(6):425-432. [PubMed 6861633]
  120. Mazuski JE, Tessier JM, May AK, et al. The Surgical Infection Society revised guidelines on the management of intra-abdominal infection. Surg Infect (Larchmt). 2017;18(1):1-76. [PubMed 28085573]
  121. McCracken GH Jr, Mize SG. A controlled study of intrathecal antibiotic therapy in gram-negative enteric meningitis of infancy. Report of the neonatal meningitis cooperative study group. J Pediatr. 1976;89(1):66-72. [PubMed 778366]
  122. McCracken GH Jr, Mize SG, Threlkeld N. Intraventricular gentamicin therapy in gram-negative bacillary meningitis of infancy. Report of the Second Neonatal Meningitis Cooperative Study Group. Lancet. 1980;1(8172):787-791. [PubMed 6102677]
  123. McDade EJ, Wagner JL, Moffett BS, et al. Once-daily gentamicin dosing in pediatric patients without cystic fibrosis. Pharmacotherapy. 2010;30(3):248-253. [PubMed 20180608]
  124. McNamara DR, Nafziger AN, Menhinick AM, Bertino JS Jr. A dose-ranging study of gentamicin pharmacokinetics: implications for extended interval aminoglycoside therapy. J Clin Pharmacol. 2001;41(4):374-377. doi:10.1177/00912700122010221 [PubMed 11304893]
  125. Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infection: 2009 update by the Infectious Diseases Society of America [published corrections appear in Clin Infect Dis. 2010;50(7):1079; Clin Infect Dis. 2010;50(3):457]. Clin Infect Dis. 2009;49(1):1-45. doi: 10.1086/599376. [PubMed 19489710]
  126. Miller MM, Burton ME, Johnson PN, Miller JL. "Once-daily" versus "extended-interval" administration of aminoglycosides in neonates: need for standard terminology. Am J Health Syst Pharm. 2014;71(24):2108-2109. [PubMed 25465576]
  127. Moehring R, Anderson DJ. Gram-negative bacillary bacteremia in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  128. Moffett BS, Kam C, Galati M, et al. The "ideal" body weight for pediatric gentamicin dosing in the era of obesity: A population pharmacokinetic analysis. Ther Drug Monit. 2018;40(3):322-329. doi: 10.1097/FTD.0000000000000505 [PubMed 29521784]
  129. Moore RD, Lietman PS, Smith CR. Clinical response to aminoglycoside therapy: importance of the ratio of peak concentration to minimal inhibitory concentration. J Infect Dis. 1987;155(1):93-99. doi:10.1093/infdis/155.1.93 [PubMed 3540140]
  130. Mueller EW, Boucher BA. The use of extended-interval aminoglycoside dosing strategies for the treatment of moderate-to-severe infections encountered in critically ill surgical patients. Surg Infect (Larchmt). 2009;10(6):563-570. doi:10.1089/sur.2007.080 [PubMed 20035611]
  131. Murray BE. Treatment of enterococcal infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 8, 2020.
  132. Murray KL, Wright D, Laxton B, Miller KM, Meyers J, Englebright J. Implementation of standardized pediatric i.v. medication concentrations. Am J Health Syst Pharm. 2014;71(17):1500-1508. [PubMed 25147175]
  133. Nelson CA, Meaney-Delman D, Fleck-Derderian S, Cooley KM, Yu PA, Mead PS; contributors. Antimicrobial treatment and prophylaxis of plague: recommendations for naturally acquired infections and bioterrorism response. MMWR Recomm Rep. 2021;70(3):1-27. doi:10.15585/mmwr.rr7003a1 [PubMed 34264565]
  134. Nezic L, Derungs A, Bruggisser M, Tschudin-Sutter S, Krähenbühl S, Haschke M. Therapeutic drug monitoring of once daily aminoglycoside dosing: comparison of two methods and investigation of the optimal blood sampling strategy. Eur J Clin Pharmacol. 2014;70(7):829-837. doi: 10.1007/s00228-014-1680-3. [PubMed 24756148]
  135. Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW, Quintiliani R. Experience with a once-daily aminoglycoside program administered to 2,184 adult patients. Antimicrob Agents Chemother. 1995;39(3):650-655. [PubMed 7793867]
  136. Nielsen EI, Cars O, Friberg LE. Pharmacokinetic/pharmacodynamic (PK/PD) indices of antibiotics predicted by a semimechanistic PKPD model: a step toward model-based dose optimization. Antimicrob Agents Chemother. 2011;55(10):4619-4630. doi:10.1128/AAC.00182-11 [PubMed 21807983]
  137. Norris AH, Shrestha NK, Allison GM, et al. 2018 Infectious Diseases Society of America clinical practice guideline for the management of outpatient parenteral antimicrobial therapy. Clin Infect Dis. 2019;68(1):e1-e35. [PubMed 30423035]
  138. Ohler KH, Menke JA, and Fuller L, "Use of Higher Dose Extended Interval Aminoglycosides in a Neonatal Intensive Care Unit," Am J Perinatol, 2000, 17(6):285-90. [PubMed 11144309]
  139. Olaison L, Schadewitz K; Swedish Society of Infectious Diseases Quality Assurance Study Group for Endocarditis. Enterococcal endocarditis in Sweden, 1995-1999: can shorter therapy with aminoglycosides be used? Clin Infect Dis. 2002;34(2):159-166. [PubMed 11740702]
  140. Otchwemah R, Grams V, Tjardes T, et al. Bacterial contamination of open fractures - pathogens, antibiotic resistances and therapeutic regimes in four hospitals of the trauma network Cologne, Germany. Injury. 2015;46(suppl 4):S104-S108. doi:10.1016/S0020-1383(15)30027-9 [PubMed 26542854]
  141. Pagkalis S, Mantadakis E, Mavros MN, Ammari C, Falagas ME. Pharmacological considerations for the proper clinical use of aminoglycosides. Drugs. 2011;71(17):2277-2294. doi: 10.2165/11597020-000000000-00000. [PubMed 22085385]
  142. Pai MP, Bearden DT. Antimicrobial dosing considerations in obese adult patients. Pharmacotherapy. 2007;27(8):1081-1091. doi:10.1592/phco.27.8.1081 [PubMed 17655508]
  143. Pai MP, Rodvold KA. Aminoglycoside dosing in patients by kidney function and area under the curve: the Sawchuk-Zaske dosing method revisited in the era of obesity. Diagn Microbiol Infect Dis. 2014;78(2):178-187. doi:10.1016/j.diagmicrobio.2013.10.011 [PubMed 24268292]
  144. Panidis D, Markantonis SL, Boutzouka E, Karatzas S, Baltopoulos G. Penetration of gentamicin into the alveolar lining fluid of critically ill patients with ventilator-associated pneumonia. Chest. 2005;128(2):545-552. doi:10.1378/chest.128.2.545 [PubMed 16100136]
  145. Penn RL. Tularemia: clinical manifestations, diagnosis, treatment, and prevention. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed December 10, 2020.
  146. Pennington JE, Reynolds HY. Pharmacokinetics of gentamicin sulfate in bronchial secretions. J Infect Dis. 1975;131(2):158-162. doi:10.1093/infdis/131.2.158 [PubMed 804018]
  147. Petejova N, Zahalkova J, Duricova J, et al. Gentamicin pharmacokinetics during continuous venovenous hemofiltration in critically ill septic patients. J Chemother. 2012;24(2):107-112. doi:10.1179/1120009X12Z.0000000006 [PubMed 22546767]
  148. Pickering LK, Ericsson CD, Ruiz-Palacios G, Blevins J, Miner ME. Intraventricular and parenteral gentamicin therapy for ventriculitis in children. Am J Dis Child. 1978;132(5):480-483. doi:10.1001/archpedi.1978.02120300040007 [PubMed 645674]
  149. Pintado V, Cabellos C, Moreno S, Meseguer MA, Ayats J, Viladrich PF. Enterococcal meningitis: a clinical study of 39 cases and review of the literature. Medicine (Baltimore). 2003;82(5):346-364. [PubMed 14530784]
  150. Popović J, Grujić Z, and Sabo A, "Influence of Pregnancy on Ceftriaxone, Cefazolin and Gentamicin Pharmacokinetics in Caesarean vs. Non-pregnant Sectioned Women," J Clin Pharm Ther, 2007, 32(6):595-602. [PubMed 18021337]
  151. Prescott WA Jr, Nagel JL. Extended-interval once-daily dosing of aminoglycosides in adult and pediatric patients with cystic fibrosis. Pharmacotherapy. 2010;30(1):95-108. doi:10.1592/phco.30.1.95 [PubMed 20030477]
  152. Raju IT, Solanki R, Patnaik AN, Barik RC, Kumari NR, Gulati AS. Brucella endocarditis - a series of five case reports. Indian Heart J. 2013;65(1):72-77. doi:10.1016/j.ihj.2012.12.017 [PubMed 23438616]
  153. Rea RS, Capitano B, Bies R, Bigos KL, Smith R, Lee H. Suboptimal aminoglycoside dosing in critically ill patients. Ther Drug Monit. 2008;30(6):674-681. doi: 10.1097/FTD.0b013e31818b6b2f. [PubMed 19057371]
  154. Reber KM, Nankervis CA. Necrotizing enterocolitis: preventative strategies. Clin Perinatol. 2004;31(1):157-167. [PubMed 15183664]
  155. Refer to manufacturer's labeling.
  156. Regamey C, Gordon RC, Kirby WM. Comparative pharmacokinetics of tobramycin and gentamicin. Clin Pharmacol Ther. 1973;14(3):396-403. [PubMed 4698568]
  157. Reimche LD, Rooney, ME, Hindmarsh KW, et al, “An Evaluation of Gentamicin Dosing According to Renal Function in Neonates With Suspected Sepsis,” Am J Perinatol, 1987, 4(3):262-5. [PubMed 3606761]
  158. Roberts JA, Field J, Visser A, et al. Using population pharmacokinetics to determine gentamicin dosing during extended daily diafiltration in critically ill patients with acute kidney injury. Antimicrob Agents Chemother. 2010;54(9):3635-3640. doi:10.1128/AAC.00222-10 [PubMed 20547809]
  159. Robinson RF, Nahata MC. Safety of intravenous bolus administration of gentamicin in pediatric patients. Ann Pharmacother. 2001;35(11):1327-1331. [PubMed 11724077]
  160. Rodvold KA, George JM, Yoo L. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antibacterial agents. Clin Pharmacokinet. 2011;50(10):637-664. doi:10.2165/11594090-000000000-00000 [PubMed 21895037]
  161. Rolain JM, Brouqui P, Koehler JE, Maguina C, Dolan MJ, Raoult D. Recommendations for treatment of human infections caused by Bartonella species. Antimicrob Agents Chemother. 2004;48(6):1921-1933. [PubMed 15155180]
  162. Ross JDC, Brittain C, Cole M, et al; G-ToG trial team. Gentamicin compared with ceftriaxone for the treatment of gonorrhoea (G-ToG): a randomised non-inferiority trial [published correction appears in Lancet. 2019;393(10191):2590]. Lancet. 2019;393(10190):2511-2520. doi: 10.1016/S0140-6736(18)32817-4. [PubMed 31056291]
  163. Roy C, Gray C, Ruda L, Bell A, Bolt J. High-dose, extended-interval gentamicin and tobramycin for pediatric inpatients: a survey of Canadian hospital pharmacists. Can J Hosp Pharm. 2016;69(5):367-375. [PubMed 27826154]
  164. Rughoo L, Bourguignon L, Maire P, Ducher M. Study of relationship between volume of distribution and body weight application to amikacin. Eur J Drug Metab Pharmacokinet. 2014;39(2):87-91. doi:10.1007/s13318-013-0160-y [PubMed 24599705]
  165. Russell GV Jr, King C, May CG, Pearsall AW 4th. Once daily high-dose gentamicin to prevent infection in open fractures of the tibial shaft: a preliminary investigation. South Med J. 2001;94(12):1185-1191. [PubMed 11811857]
  166. Ryanto S, Wong M, Czarniak P, et al. The use of initial dosing of gentamicin in the management of pyelonephritis/urosepsis: a retrospective study. PLoS One. 2019;14(1):e0211094. doi:10.1371/journal.pone.0211094 [PubMed 30673763]
  167. Rybak MJ, Abate BJ, Kang SL, Ruffing MJ, Lerner SA, Drusano GL. Prospective evaluation of the effect of an aminoglycoside dosing regimen on rates of observed nephrotoxicity and ototoxicity. Antimicrob Agents Chemother. 1999;43(7):1549-1555. [PubMed 10390201]
  168. Safi KH, Damiani JM, Sturza J, Nasr SZ. Extended-interval aminoglycoside use in cystic fibrosis exacerbation in children and young adults: a prospective quality improvement project. Glob Pediatr Health. 2016;3:2333794X16635464. [PubMed 27336007]
  169. Schmidt GA, Mandel J. Evaluation and management of suspected sepsis and septic shock in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  170. Shankar SM, Jew RK, Bickert BM, Cavalieri GE, Bell LM, Lange BJ. Pharmacokinetics of single daily dose gentamicin in children with cancer. J Pediatr Hematol Oncol. 1999;21(4):284-288. doi:10.1097/00043426-199907000-00008 [PubMed 10445890]
  171. Shevchuk YM and Taylor DM, “Aminoglycoside Volume of Distribution in Pediatric Patients,” DICP, 1990, 24(3):273-6. [PubMed 2316235]
  172. Shields RK, Clancy CJ, Press EG, Nguyen MH. Aminoglycosides for treatment of bacteremia due to carbapenem-resistant Klebsiella pneumoniae. Antimicrob Agents Chemother. 2016;60(5):3187-3192. doi: 10.1128/AAC.02638-15. [PubMed 26926642]
  173. Skalsky K, Yahav D, Bishara J, Pitlik S, Leibovici L, Paul M. Treatment of human brucellosis: systematic review and meta-analysis of randomised controlled trials. BMJ. 2008;336(7646):701-704. doi: 10.1136/bmj.39497.500903.25. [PubMed 18321957]
  174. Solera J, Geijo P, Largo J, et al; Grupo de Estudio de Castilla-la Mancha de Enfermedades Infecciosas. A randomized, double-blind study to assess the optimal duration of doxycycline treatment for human brucellosis. Clin Infect Dis. 2004;39(12):1776-1782. doi:10.1086/426024 [PubMed 15578399]
  175. Solera J, Lozano E, Martínez-Alfaro E, Espinosa A, Castillejos ML, Abad L. Brucellar spondylitis: review of 35 cases and literature survey. Clin Infect Dis. 1999;29(6):1440-1449. doi:10.1086/313524 [PubMed 10585793]
  176. 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. Surg Infect (Larchmt). 2010;11(1):79-109. [PubMed 20163262]
  177. Sorger JI, Kirk PG, Ruhnke CJ, et al. Once daily, high dose versus divided, low dose gentamicin for open fractures. Clin Orthop Relat Res. 1999;(366):197-204. doi:10.1097/00003086-199909000-00026 [PubMed 10627736]
  178. Southgate WM, DiPiro JT, and Robertson AF, "Pharmacokinetics of Gentamicin in Neonates on Extracorporeal Membrane Oxygenation," Antimicrob Agents Chemother, 1989, 33(6):817-9. [PubMed 2764529]
  179. Sowinski KM, Magner SJ, Lucksiri A, Scott MK, Hamburger RJ, Mueller BA. Influence of hemodialysis on gentamicin pharmacokinetics, removal during hemodialysis, and recommended dosing. Clin J Am Soc Nephrol. 2008;3(2):355-361. doi:10.2215/CJN.02920707 [PubMed 18235142]
  180. Spach DH, Kaplan SL. Treatment of cat scratch disease. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 20, 2022a.
  181. Spach DH. Diagnosis, treatment, and prevention of Bartonella infections in HIV-infected patients. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 2, 2022b.
  182. Spach DH. Clinical features, diagnosis, and treatment of Bartonella quintana infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 2, 2022c.
  183. Spach DH. Endocarditis caused by Bartonella. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 2, 2022d.
  184. St. Cyr S, Barbee L, Workowski KA, et al. Update to CDC's treatment guidelines for gonococcal infection, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(50):1911-1916. doi:10.15585/mmwr.mm6950a6 [PubMed 33332296]
  185. Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America [published online June 18, 2014]. Clin Infect Dis. 2014;59(2):e10-52. doi: 10.1093/cid/ciu296. [PubMed 24947530]
  186. Stout J. Clinical manifestations, diagnosis, and treatment of plague (Yersinia pestis infection). Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed March 17, 2022.
  187. Szeto CC, Li PK. Concerns regarding inconsistencies within and between ISPD recommendations for peritonitis and catheter-related infections-in reply. Perit Dial Int. 2018;38(4):311-312. doi: 10.3747/pdi.2018.00046. [PubMed 29987069]
  188. Tamma PD, Aitken SL, Bonomo RA, Mathers AJ, van Duin D, Clancy CJ. Infectious Diseases Society of America Antimicrobial Resistant Treatment Guidance: Gram-Negative Bacterial Infections. Clin Infect Dis. Published online October 27, 2020. [PubMed 33106864]
  189. Tita ATN. Intraamniotic infection (clinical chorioamnionitis). Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed July 27, 2022.
  190. Touw DJ, Westerman EM, Sprij AJ. Therapeutic drug monitoring of aminoglycosides in neonates. Clin Pharmacokinet. 2009;48(2):71-88. [PubMed 19271781]
  191. Townsend PL, Fink MP, Stein KL, et al, “Aminoglycoside Pharmacokinetics: Dosage Requirements and Nephrotoxicity in Trauma Patients,” Crit Care Med, 1989, 17(2):154-7. [PubMed 2914447]
  192. Traynor AM, Nafziger AN, Bertino JS Jr. Aminoglycoside dosing weight correction factors for patients of various body sizes. Antimicrob Agents Chemother. 1995;39(2):545-548. doi:10.1128/aac.39.2.545 [PubMed 7726530]
  193. Trotman RL, Williamson JC, Shoemaker DM, et al, “Antibiotic Dosing in Critically Ill Adult Patients Receiving Continuous Renal Replacement Therapy,” Clin Infect Dis, 2005, 41(8):1159-66. [PubMed 16163635]
  194. Tunkel AR. Treatment of bacterial meningitis caused by specific pathogens in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 9, 2019.
  195. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the management of bacterial meningitis. Clin Infect Dis. 2004;39(9):1267-1284. [PubMed 15494903]
  196. Tunkel AR, Hasbun R, Bhimraj A, et al. 2017 Infectious Diseases Society of America's clinical practice guidelines for healthcare-associated ventriculitis and meningitis. Clin Infect Dis. 2017;64(6):e34-e65. [PubMed 28203777]
  197. Udy AA, Roberts JA, Boots RJ, Paterson DL, Lipman J. Augmented renal clearance: implications for antibacterial dosing in the critically ill. Clin Pharmacokinet. 2010;49(1):1-16. doi:10.2165/11318140-000000000-00000 [PubMed 20000886]
  198. Urban AW, Craig WA. Daily dosage of aminoglycosides. Curr Clin Top Infect Dis. 1997;17:236-255. [PubMed 9189668]
  199. US Department of Health and Human Services (HHS) Panel on Opportunistic Infections in Adults and Adolescents With HIV. Guidelines for the prevention and treatment of opportunistic infections in adults and adolescents with HIV: 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. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/Adult_OI.pdf. Updated February 17, 2022. Accessed January 31, 2022.
  200. Valitalo PA, van den Anker JN, Allegaert K, et al. Novel model-based dosing guidelines for gentamicin and tobramycin in preterm and term neonates. J Antimicrob Chemother. 2015;70(7):2074-2077. [PubMed 25766737]
  201. van Donge T, Pfister M, Bielicki J, et al. Quantitative Analysis of Gentamicin Exposure in Neonates and Infants Calls into Question Its Current Dosing Recommendations. Antimicrob Agents Chemother. 2018;62(4):e02004-17. [PubMed 29358294]
  202. van Maarseveen EM, Sprij A, Touw DJ. Extended-interval dosing of gentamicin aiming for a drug-free period in neonates: a prospective cohort study. Ther Drug Monit. 2016;38(3):402-406. [PubMed 26836810]
  203. Van Meter DJ, Corriveau M, Ahern JW, et al, "A Survey of Once-Daily Dosage Tobramycin Therapy in Patients With Cystic Fibrosis," Pediatr Pulmonol, 2009, 44(4):325-9. [PubMed 19330772]
  204. Varese LA, Grazioli F, Viretto A, Antoniola P. Single-dose (bolus) therapy with gentamicin in management of urinary tract infection. International Journal of Pediatric Nephrology. 1980;1(2):104-105.
  205. Vercaigne LM, Zelenitsky SA, Findlay I, Bernstein K, Penner SB. An in vitro evaluation of the antibiotic/heparin lock to sterilize central venous haemodialysis catheters. J Antimicrob Chemother. 2002;49(4):693-696. [PubMed 11909847]
  206. Vermont Oxford Network (VON). Neonatal drug concentrations. Available at: https://public.vtoxford.org/neonatal-drug-concentrations. Updated November 2022. Accessed August 10, 2023.
  207. Verpooten GA, Giuliano RA, Verbist L, Eestermans G, De Broe ME. Once-daily dosing decreases renal accumulation of gentamicin and netilmicin. Clin Pharmacol Ther. 1989;45(1):22-27. doi:10.1038/clpt.1989.4 [PubMed 2910634]
  208. Wallace AW, Jones M, Bertino JS Jr. Evaluation of four once-daily aminoglycoside dosing nomograms. Pharmacotherapy. 2002;22(9):1077-1083. doi:10.1592/phco.22.13.1077.33529 [PubMed 12222541]
  209. Warady BA, Bakkaloglu S, Newland J, et al, "Consensus Guidelines for the Prevention and Treatment of Catheter-Related Infections and Peritonitis in Pediatric Patients Receiving Peritoneal Dialysis: 2012 Update," Perit Dial Int, 2012, (32 Suppl 2):S32-86. [PubMed 22851742]
  210. Wassil SK, Fox KM, White JW. Once daily dosing of aminoglycosides in pediatric cystic fibrosis patients: a review of the literature. J Pediatr Pharmacol Ther. 2008;13(2):68-75. [PubMed 23055867]
  211. Wie SH, Kim HW, Chang UI. Effects of gentamicin monotherapy for the initial treatment of community-onset complicated non-obstructive acute pyelonephritis due to Enterobacteriaceae in elderly and non-elderly women. Clin Microbiol Infect. 2014;20(11):1211-1218. doi:10.1111/1469-0691.12711 [PubMed 24909648]
  212. Wiesenfeld HC. Pelvic inflammatory disease: treatment in adults and adolescents. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed April 11, 2022.
  213. 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(15):1736-54. Available at http://circ.ahajournals.org/cgi/reprint/CIRCULATIONAHA.106.183095v1 [PubMed 17446442]
  214. Workowski KA, Bachmann LH, Chan PA; Centers for Disease Control and Prevention (CDC). Sexually transmitted infections treatment guidelines, 2021. MMWR Recomm Rep. 2021;70(4):1-187. doi:10.15585/mmwr.rr7004a1 [PubMed 34292926]
  215. Workowski KA, Bolan GA; Centers for Disease Control and Prevention (CDC). Sexually transmitted diseases treatment guidelines, 2015 [published correction appears in MMWR Recomm Rep. 2015;64(33):924]. MMWR Recomm Rep. 2015;64(RR-03):1-137. [PubMed 26042815]
  216. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the eleventh WHO model list of essential drugs. 2002. Available at http://www.who.int/maternal_child_adolescent/documents/55732/en/
  217. World Health Organization (WHO). Brucellosis in humans and animals. Published 2006. http://www.who.int/csr/resources/publications/Brucellosis.pdf.
  218. World Health Organization (WHO). WHO guidelines on tularaemia. http://whqlibdoc.who.int/publications/2007/9789241547376_eng.pdf. Published 2007. Accessed June 23, 2017.
  219. Xuan D, Nicolau DP, Nightingale CH. Population pharmacokinetics of gentamicin in hospitalized patients receiving once-daily dosing. Int J Antimicrob Agents. 2004;23(3):291-295. doi:10.1016/j.ijantimicag.2003.07.010 [PubMed 15164971]
  220. Zaske DE. Aminoglycosides. In: Evans WE, Schentag JJ, Jusko WJ, eds. Applied Pharmacokinetics. 2nd ed. Spokane, WA: Applied Therapeutics Inc; 1986, p. 362.
  221. Zaske DE, Cipolle RJ, Rotschafer JC, Solem LD, Mosier NR, Strate RG. Gentamicin pharmacokinetics in 1,640 patients: method for control of serum concentrations. Antimicrob Agents Chemother. 1982;21(3):407-411. [PubMed 7103444]
  222. Zelenitsky SA, Harding GK, Sun S, Ubhi K, Ariano RE. Treatment and outcome of Pseudomonas aeruginosa bacteraemia: an antibiotic pharmacodynamic analysis. J Antimicrob Chemother. 2003;52(4):668-674. doi:10.1093/jac/dkg403 [PubMed 12951354]
  223. Zobell JT, Epps K, Kittell F, et al. Tobramycin and beta-lactam antibiotic use in cystic fibrosis exacerbations: a pharmacist approach. J Pediatr Pharmacol Ther. 2016;21(3):239-246. [PubMed 27453702]
Topic 15997 Version 326.0

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟