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

Amikacin (systemic): Drug information

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

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

Neurotoxicity, manifested as vestibular and permanent bilateral auditory ototoxicity, can occur in patients with preexisting renal damage and in patients with normal renal function treated at higher doses and/or periods longer than those recommended. The risk of aminoglycoside-induced ototoxicity is greater in patients with renal damage. High frequency deafness usually occurs first and can be detected only by audiometric testing. Vertigo may occur and may be evidence of vestibular injury. Other manifestations of neurotoxicity may include numbness, skin tingling, muscle twitching, and convulsions. The risk of hearing loss due to aminoglycosides increases with the degree of exposure to either high peak or high trough serum concentrations. Patients developing cochlear damage may not have symptoms during therapy to warn them of developing eighth-nerve toxicity, and total or partial irreversible bilateral deafness may occur after the drug has been discontinued. Aminoglycoside-induced ototoxicity is usually irreversible. Patients treated with parenteral aminoglycosides should be under close clinical observation because of the potential ototoxicity associated with their use. Safety for treatment periods which are longer than 14 days has not been established.

Nephrotoxicity:

Aminoglycosides are potentially nephrotoxic. The risk of nephrotoxicity is greater in patients with impaired renal function and in those who receive high doses or prolonged therapy. Patients treated with parenteral aminoglycosides should be under close clinical observation because of the potential nephrotoxicity associated with their use. Safety for treatment periods which are longer than 14 days has not been established.

Neuromuscular blockade:

Neuromuscular blockade and respiratory paralysis have been reported following parenteral injection, topical instillation (as in orthopedic and abdominal irrigation or in local treatment of empyema), and following oral use of aminoglycosides. The possibility of these phenomena should be considered if aminoglycosides are administered by any route, especially in patients receiving anesthetics, neuromuscular blocking agents such as tubocurarine, succinylcholine, decamethonium, or in patients receiving massive transfusions of citrate-anticoagulated blood. If blockage occurs, calcium salts may reverse these phenomena, but mechanical respiratory assistance may be necessary.

Monitoring:

Renal and eighth-nerve function should be closely monitored especially in patients with known or suspected renal impairment at the onset of therapy and also in those whose renal function is initially normal but who develop signs of renal dysfunction during therapy. Serum concentrations of amikacin should be monitored when feasible to assure adequate levels and to avoid potentially toxic levels and prolonged peak concentrations above 35 mcg/mL. Urine should be examined for decreased specific gravity, increased excretion of proteins, and the presence of cells or casts. Blood urea nitrogen, serum creatinine, or creatinine clearance should be measured periodically. Serial audiograms should be obtained where feasible in patients old enough to be tested, particularly high-risk patients. Evidence of ototoxicity (dizziness, vertigo, tinnitus, roaring in the ears, and hearing loss) or nephrotoxicity requires discontinuation of the drug or dosage adjustment.

Concurrent therapy:

Concurrent and/or sequential systemic, oral, or topical use of other neurotoxic or nephrotoxic products, particularly bacitracin, cisplatin, amphotericin B, cephaloridine, paromomycin, viomycin, polymyxin B, colistin, vancomycin, or other aminoglycosides should be avoided. Other factors that may increase risk of toxicity are advanced age and dehydration.

The concurrent use of amikacin with potent diuretics (ethacrynic acid, or furosemide) should be avoided because diuretics by themselves may cause ototoxicity. In addition, when administered intravenously, diuretics may enhance aminoglycoside toxicity by altering antibiotic concentrations in serum and tissue.

Brand Names: Canada
  • Erfa-Amikacin;
  • VPI-Amikacin
Pharmacologic Category
  • Antibiotic, Aminoglycoside
Dosing: Adult

Dosing guidance:

Dosing: For patients who are underweight (ie, total body weight [TBW] < ideal body weight [IBW]), calculate 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 × IBW), use adjusted body weight ([0.4 × (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:

Conventional/traditional dosing: IM, IV: 5 mg/kg every 8 hours or 7.5 mg/kg every 12 hours. Target peak concentration depends on indication, site of infection, and minimum inhibitory concentration (MIC) of pathogen; in general, adjust dose to achieve peak of 20 to 40 mg/L. Target trough concentrations should be <8 mg/L; ideal target trough is 1 to 4 mg/L (Ref).

High - dose extended-interval dosing: IV: 15 to 20 mg/kg once daily; use with caution in patients with CrCl <40 mL/minute (Ref). Some experts recommend doses up to 30 mg/kg once daily in critically ill patients (Ref). Adjust amikacin dose and interval to achieve a peak concentration of ~40 mg/L (60 mg/L may be needed in critically ill patients) and trough concentration <2 mg/L (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 on dialysis, pregnancy) (Ref); some experts prefer traditional intermittent dosing in such populations (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: 15 to 30 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. Amikacin should not be used as monotherapy (Ref).

Cerebrospinal fluid shunt infection

Cerebrospinal fluid (CSF) shunt infection (susceptible gram-negative organisms):

IV: 5 mg/kg every 8 hours in combination with other appropriate agents (Ref).

Intraventricular/intrathecal (adjunct to systemic therapy; use a preservative-free preparation): 5 to 50 mg/day; usual dose: 30 mg/day (Ref). Some experts recommend adjusting dosage and administration interval based on CSF amikacin concentrations (goal: 10 to 20 times MIC of causative organism), ventricle size, and daily output from ventricular drain (Ref). When intraventricular amikacin 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). Note: Intraventricular/intrathecal administration is generally reserved for use in patients who fail parenteral therapy despite removal of CSF shunt or when CSF shunt cannot be removed (Ref).

Cystic fibrosis, acute pulmonary exacerbation

Cystic fibrosis, acute pulmonary exacerbation (off-label use): IV: 30 to 35 mg/kg once daily in combination with other appropriate agents; adjust dose and interval to achieve a target peak of 80 to 120 mg/L and trough <1 mg/L (Ref).

Meningitis, bacterial

Meningitis, bacterial (susceptible gram-negative organisms): IV: 5 mg/kg every 8 hours in combination with other appropriate agents (Ref).

Mycobacterial infection

Mycobacterial (nontuberculous) infection:

M. avium complex (off-label use):

Disseminated disease in patients with HIV, adjunct therapy: IV: 10 to 15 mg/kg daily as part of an appropriate combination regimen. Note: Some experts recommend addition of amikacin in patients with high mycobacterial loads (ie, >2 log CFU/mL of blood) or in the absence of effective antiretroviral therapy (Ref).

Pulmonary disease (severe nodular/bronchiectatic or cavitary disease) (adjunctive agent): IV: 10 to 15 mg/kg once daily or 15 to 25 mg/kg 3 times weekly for first 2 to 3 months as part of an appropriate combination regimen (Ref).

Rapidly growing mycobacteria (eg, M. abscessus) (off-label use):

Initial therapy: IV: 10 to 15 mg/kg once daily or 15 to 25 mg/kg 3 times weekly as part of an appropriate combination regimen. The optimal duration of therapy is unknown, but generally the duration of parenteral therapy is ≤12 weeks depending on pathogen, severity of infection, tolerability, and other patient-specific factors, followed by long-term oral maintenance therapy; consult an infectious diseases specialist for specific recommendations (Ref).

Maintenance therapy: Inhalation for nebulization (using injection solution): 250 to 500 mg once or twice daily as part of an appropriate combination regimen; consult an infectious diseases specialist for specific recommendations including duration (Ref).

Nocardiosis, severe

Nocardiosis, severe (off-label use):

Note: Due to concerns for resistance, susceptibility testing should be performed on isolates (Ref).

IV: 10 to 15 mg/kg once daily as part of an appropriate combination regimen (Ref). Prolonged treatment is required (range 6 months to ≥1 year [at least several weeks of parenteral therapy followed by oral therapy]) (Ref). Consult an infectious diseases specialist for specific treatment recommendations.

Plague, treatment

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

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

IV, IM: 15 to 20 mg/kg once daily for 7 to 14 days and for at least a few days after clinical resolution (Ref).

Pneumonia, hospital-acquired or ventilator-associated

Pneumonia, hospital-acquired or ventilator-associated (alternative agent):

Note: Some experts reserve for patients with risk for multidrug-resistant pathogens (Ref).

IV: 15 to 20 mg/kg once daily in combination with a second gram-negative agent; once culture and susceptibility results are available, can generally discontinue amikacin and use a single agent with documented activity (Ref). Note: Avoid use of amikacin monotherapy (Ref).

Inhalation for nebulization (using injectable solution): 400 to 500 mg every 12 hours in combination with IV antimicrobial therapy. Note: Efficacy is uncertain and bronchospasm may limit use (Ref).

Sepsis or septic shock, adjunctive empiric gram-negative coverage

Sepsis or septic shock, adjunctive empiric gram-negative coverage:

Note: Some experts reserve for patients at high risk for multidrug resistant gram-negative pathogens (Ref).

IV: 15 to 30 mg/kg once daily in combination with a second gram-negative agent; once culture and susceptibility tests are available, can generally discontinue and use a single agent with documented activity. Amikacin should not be used as monotherapy (Ref).

Tuberculosis, drug resistant

Tuberculosis, drug resistant (alternative agent) (off-label use):

Note: Expert consultation for optimal regimen and duration of treatment is advised.

IM, IV: 15 mg/kg once daily or 25 mg/kg 3 times weekly in combination with additional appropriate antituberculosis agents (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: IV:

Note: Use with caution in patients with CrCl <40 mL/minute (Ref), although 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).

Initial dose: 15 to 20 mg/kg; up to 30 mg/kg once daily in critically ill patients. 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 (ie, critical illness, pregnancy, etc) (Ref). Also refer to institutional-specific policies. The following recommendations may serve as a general guideline after the initial dose:

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

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

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

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

Conventional/traditional dosing: IM, IV:

Note: High-dose, extended-interval dosing is generally preferred for treatment of gram-negative infections.

Regimens may vary based on individualized pharmacokinetic calculations and pharmacodynamic targets; also refer to institutional-specific policies. The following recommendations are expert opinion derived from Leroy 1978:

Amikacin Dose Adjustments for Altered Kidney Function

CrCl

If the usual indication-specific dose is 7.5 mg/kg every 12 hours or 5 mg/kg every 8 hours

≥60 mL/minute

No dosage adjustment necessary.

40 to <60 mL/minute

5 to 7.5 mg/kg every 12 hours.

20 to <40 mL/minute

5 to 7.5 mg/kg every 24 hours.

<20 mL/minute

5 to 7.5 mg/kg once; subsequent doses should be based on amikacin 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: 20 to 30 mg/kg once daily for known/suspected sepsis; adjust dose and/or interval based on close monitoring of amikacin serum concentrations and individualized pharmacokinetic calculations and pharmacodynamic targets; use of nomograms for dosage adjustment are not recommended in this population (expert opinion derived from Aréchiga-Alvarado 2020).

Hemodialysis, intermittent (thrice weekly):

Dialyzable (~20% to 65% (Ref) dependent on filter and duration): Note: Postdialysis concentrations should be drawn ≥2 and up to 4 hours after hemodialysis to allow for redistribution (Ref).

IM, IV: 5 to 12.5 mg/kg/dose 3 times weekly after dialysis on dialysis days (Ref). Redose when prehemodialysis amikacin concentration <10 mg/L or when posthemodialysis amikacin concentration <6 to 8 mg/L (Ref).

Peritoneal dialysis: IM, IV: Initial: 5 to 12.5 mg/kg/dose (depending on infection site, severity, and susceptibility of infecting organisms) every 48 to 72 hours; adjust dose and/or interval based on amikacin 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) and minimal residual kidney function 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: IV: Initial: 15 to 25 mg/kg (depending on infection site, severity, and susceptibility of infecting organisms) every 48 hours. Adjust dose and/or interval, as needed, based upon amikacin serum concentrations (Ref).

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. The following dosing assumes daily use of PIRRT.

IV: Initial: 15 to 25 mg/kg (depending on infection site, severity, and susceptibility of infecting organisms) every 48 hours. Adjust dose and/or interval, as needed, based upon amikacin serum concentrations. 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.

Dosing: Obesity: Adult

The recommendations for dosing in patients with obesity are based upon the best available evidence and clinical expertise. Senior Editorial Team: Jeffrey F. Barletta, PharmD, FCCM; Manjunath P. Pai, PharmD, FCP; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC.

Class 1, 2, or 3 obesity (BMI ≥30 kg/m2):

IV: Use adjusted body weight for initial weight-based dosing when targeting Cmax/minimum inhibitory concentration (MIC) goals with either loading dose equation (based on target aminoglycoside concentration and estimated Vd) or mg/kg approach and when estimating kidney function with Cockcroft-Gault (CrCl) (Ref). Alternatively, use adjusted body weight to estimate CrCl and aminoglycoside dosing with the Bayesian approach when targeting AUC/MIC goals (Ref). Note: If aminoglycoside therapy is continued, use Cmax/MIC or AUC/MIC goals to optimize therapy, especially in the critically ill where weight and kidney function may be poor surrogates of Vd and clearance (Ref).

Rationale for recommendations: Aminoglycosides are hydrophilic medications with a low Vd and clearance that is proportional to GFR. Pharmacokinetic studies have observed adjusted body weight, using a correction factor of 0.4, is the most appropriate weight metric to correct Vd in the setting of obesity. However, there is wide variation in the correction factors reported, which could lead to under- or over-dosing in clinical practice (Ref). Early use of therapeutic drug monitoring is recommended (Ref).

Dosing: Older Adult

Refer to adult dosing.

Dosing: Pediatric

(For additional information see "Amikacin (systemic): Pediatric drug information")

Note: Routes of administration may vary (including IM, IV, intraperitoneal, intrathecal, and intraventricular); dosing presented as mg/kg/dose and mg/kg/day; use caution. 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 aminoglycoside concentration monitoring is individualized based on dosing and monitoring strategy (Ref).

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

General dosing: Infants, Children, and Adolescents: Note: Extended-interval dosing is typically preferred (Ref):

Extended-interval dosing: Limited data available: IV, IM: 15 to 30 mg/kg/dose every 24 hours (Ref). Doses on the higher end of the range may be necessary in patients with critical illness, malignancy, cystic fibrosis, or burn injury, or for bacterial pathogens with minimum inhibitory concentration (MIC) ≥4 mg/L (Ref).

Conventional dosing: IV, IM: 15 to 22.5 mg/kg/day divided every 8 to 12 hours (Ref).

CNS infection

CNS infection:

Meningitis, including health care-associated meningitis: Limited data available: Infants, Children, and Adolescents: IV: 20 to 30 mg/kg/day divided every 8 hours; individualize duration based on patient characteristics, infecting bacteria, and response (Ref).

Ventriculitis (including health care-associated ventriculitis and cerebrospinal fluid [CSF] shunt infections): Limited data available:

Intraventricular/intrathecal (use a preservative-free preparation): Infants, Children, and Adolescents: 5 to 50 mg/day; usual dose: 30 mg/day (Ref). Due to the smaller CSF volume in infants, some guidelines recommend decreasing the infant dose; dosage and administration interval can also be adjusted based on CSF amikacin concentrations, ventricle size, and daily output from ventricular drain (Ref). Duration is individualized according to clinical and microbiological response (Ref).

Cystic fibrosis, acute pulmonary exacerbation

Cystic fibrosis, acute pulmonary exacerbation:

Note: Extended-interval dosing is preferred over traditional dosing. Use as part of appropriate combination therapy (Ref). Treatment duration varies and is dependent on patient-specific factors including response to therapy; typical duration is 10 to 21 days (Ref).

Extended-interval dosing: Infants, Children, and Adolescents: IV, IM: 30 to 35 mg/kg/dose every 24 hours (Ref). Higher doses (eg, 40 mg/kg/dose every 24 hours) may be necessary when MIC is ≥8 mg/L (Ref).

Traditional dosing: Infants, Children, and Adolescents: IV, IM: 10 mg/kg/dose every 8 hours (Ref).

Endocarditis, treatment

Endocarditis, treatment: Children and Adolescents: IV: 15 mg/kg/day divided every 8 to 12 hours; use in combination with other antibiotics dependent upon pathogen and source of infection (Ref).

Intra-abdominal infection, complicated

Intra-abdominal infection, complicated: Infants, Children, and Adolescents: IV: 15 to 22.5 mg/kg/day divided every 8 to 24 hours (Ref).

Mycobacterial infection

Mycobacterial (nontuberculous) infection: Limited data available:

Mycobacterium avium complex infection in persons that are HIV-exposed/-infected (adjunct therapy):

Infants and Children: IV: 15 to 30 mg/kg/day divided every 12 to 24 hours as part of an appropriate combination regimen; maximum daily dose: 1,500 mg/day; total duration of therapy for M. avium complex infection is ≥12 months (Ref).

Adolescents: IV: 10 to 15 mg/kg/dose every 24 hours as part of an appropriate combination regimen; maximum daily dose: 1,500 mg/day; total duration of therapy for M. avium complex infection is ≥12 months (Ref).

Pulmonary infection in patients with cystic fibrosis (eg, Mycobacterium abscessus, M. avium complex) (Ref): Note: Use as part of an appropriate combination regimen. The duration of parenteral therapy is generally 3 to 12 weeks depending on clinical response, followed by transition to oral and/or inhaled therapy; total treatment duration is ≥12 months after culture conversion.

Infants and Children: IV: 15 to 30 mg/kg/dose every 24 hours; maximum dose: 1,500 mg/dose.

Adolescents: IV: 10 to 15 mg/kg/dose every 24 hours; maximum dose: 1,500 mg/dose.

Tuberculosis, active; treatment

Tuberculosis, active (drug-resistant); treatment (alternative agent): Limited data available: Note: Use as part of an appropriate combination regimen; duration should be individualized based on extent of disease, rapidity of culture conversion, clinical response, and toxicity (Ref).

Infants, Children, and Adolescents: IM, IV: 15 to 20 mg/kg/dose every 24 hours (Ref).

Peritonitis

Peritonitis (CAPD): Infants, Children, and Adolescents: Intraperitoneal: Continuous: Loading dose: 25 mg per liter of dialysate; maintenance dose: 12 mg per liter (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

Infants, Children, and Adolescents: IM, IV:

The following adjustments have been recommended (Ref); Note: Renally adjusted dose recommendations are based on doses of 5 to 7.5 mg/kg/dose every 8 hours:

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

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: 5 mg/kg/dose; redose as indicated by serum concentrations.

Peritoneal dialysis (PD): 5 mg/kg/dose; redose as indicated by serum concentrations.

Continuous renal replacement therapy (CRRT): 7.5 mg/kg/dose every 12 hours, monitor serum concentrations.

Dosing: Hepatic Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling.

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified.

Frequency not defined:

Nervous system: Neurotoxicity (including muscle twitching, numbness, seizure, tingling of skin)

Otic: Auditory ototoxicity, vestibular ototoxicity

Renal: Nephrotoxicity

Respiratory: Respiratory paralysis

Postmarketing:

Cardiovascular: Hypotension

Dermatologic: Skin rash

Endocrine & metabolic: Albuminuria, hypomagnesemia

Gastrointestinal: Clostridium difficile-associated diarrhea, nausea, vomiting

Genitourinary: Azotemia, hematuria, oliguria, toxic nephrosis

Hematologic & oncologic: Anemia, eosinophilia, leukocyturia

Hypersensitivity: Drug reaction with eosinophilia and systemic symptoms (Bensaid 2012)

Nervous system: Drug fever, headache, paresthesia, tremor

Neuromuscular & skeletal: Arthralgia, state of neuromuscular blockade (Hashimoto 1978)

Renal: Acute kidney injury, casts in urine, increased serum creatinine

Contraindications

Hypersensitivity to amikacin, 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: [US Boxed Warning]: May cause neuromuscular blockade and respiratory paralysis; especially when given soon after anesthesia or muscle relaxants.

• 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 C. difficile-associated diarrhea (CDAD) and pseudomembranous colitis; CDAD has been observed >2 months postantibiotic treatment.

Disease-related concerns:

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

• Hypocalcemia: Use with caution in patients with hypocalcemia.

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

• 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.

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, bacitracin, cisplatin, amphotericin B, paromomycin, 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.

Dosage form specific issues:

• Sulfites: May contain sulfites which may cause allergic-type reactions (including anaphylaxis) as well as life-threatening or less severe asthmatic episodes in certain individuals.

Other warnings/precautions:

• Surgical irrigation: Irreversible deafness, renal failure, and death due to neuromuscular blockade have been reported following use of aminoglycosides as surgical irrigation; rapid systemic absorption occurs with topical application (except to the urinary bladder).

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.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Solution, Injection, as sulfate:

Generic: 1 g/4 mL (4 mL [DSC])

Solution, Injection, as sulfate [preservative free]:

Generic: 500 mg/2 mL (2 mL); 1 g/4 mL (4 mL)

Generic Equivalent Available: US

Yes

Pricing: US

Solution (Amikacin Sulfate Injection)

1 g/4 mL (per mL): $3.31 - $7.35

500 mg/2 mL (per mL): $3.31 - $7.36

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, as sulfate:

Generic: 250 mg/mL (2 mL); 500 mg/2 mL (2 mL)

Administration: Adult

IM: Administer IM injection in large muscle mass.

IV: Infuse over 30 to 60 minutes.

Some penicillins (eg, carbenicillin, ticarcillin, and piperacillin) have been shown to inactivate 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.

Intrathecal/Intraventricular (off-label route): Reconstitute with preservative-free diluent (NS) only to a final volume of 3 mL (Ref). When administered through a ventricular drain, clamp drain for 15 to 60 minutes before opening the drain to allow amikacin solution to equilibrate in the CSF (Ref).

Inhalation (injection formulation; off-label route): Nebulization: Use with standard jet nebulizer connected to an air compressor or ultrasonic nebulizer; administer with mouthpiece or face mask (Ref). Dilute with NS up to total volume of 4 mL (Ref).

Administration: Pediatric

Parenteral:

IM: Administer undiluted into a large muscle mass.

IV: Intermittent IV infusion: Administer by intermittent infusion over 30 to 60 minutes (Ref). Shorter infusion times of 20 minutes have been reported in neonates and slow IV bolus (eg, over 1 to 5 minutes) has also been reported in pediatric patients, including preterm and term neonates (Ref). Avoid infusing concomitantly with penicillins or cephalosporins if feasible; consult drug interactions database for more information.

Intrathecal/intraventricular: Use preservative-free preparations only. 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 solution to equilibrate in the cerebrospinal fluid (Ref).

Use: Labeled Indications

Serious infections: Treatment of serious infections (eg, bloodstream infection, bone infection, respiratory tract infection, endocarditis) due to gram-negative organisms, including Pseudomonas, Escherichia coli, Proteus, Providencia, Klebsiella, Enterobacter, Serratia, and Acinetobacter.

Use: Off-Label: Adult

Cystic fibrosis, acute pulmonary exacerbation; Mycobacterium avium complex infection; Mycobacterium (nontuberculous, rapidly growing) infection; Nocardiosis, severe; Plague (Yersinia pestis), treatment; Tuberculosis

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

Amikacin may be confused with Amicar, anakinra

Amikin may be confused with Amicar, Kineret

High alert medication:

The Institute for Safe Medication Practices (ISMP) includes this medication (intrathecal administration) among its list of drugs that 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.

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

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

Reproductive Considerations

Evaluate pregnancy status prior to treatment of multidrug-resistant tuberculosis in patients who could become pregnant. Patients who could become pregnant should use effective contraception during treatment for multidrug-resistant tuberculosis (Esmail 2018).

Pregnancy Considerations

Amikacin crosses the placenta.

Aminoglycosides may cause fetal harm if administered during pregnancy. There are several reports of total irreversible bilateral congenital deafness in children whose mothers received a different 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 intravenous amikacin may be altered. Thirty patients between 6 to 20 weeks' gestation were given a single IM dose of amikacin 7.5 mg/kg within 24 hours prior to elective hysterectomy and therapeutic abortion. Mean peak concentrations and half-life of amikacin were slightly lower than in nonpregnant adults but still within the normal therapeutic range (Bernard 1977).

Amikacin is recommended off label as part of a multiantibiotic treatment regimen of nontuberculosis Mycobacterium avium complex (MAC) in patients with cystic fibrosis in certain situations; route of administration (IV or nebulized) may be dependent upon severity and location of infection (CFF/ECFS [Floto 2016]). Use of the IV route should be reserved for life-threatening infections in pregnant patients due to the potential risk of fetal toxicity (Panchaud 2016).

Amikacin is used off label for the treatment of multidrug-resistant tuberculosis. Tuberculosis (TB) disease (active TB) is associated with adverse fetal outcomes, including intrauterine growth restriction, low birth weight, preterm birth, and perinatal death (Esmail 2018; Miele 2020), as well as adverse maternal outcomes, including increased risks for anemia and cesarean delivery. Placental transmission may rarely occur with active maternal disease (Miele 2020). The treatment of multidrug-resistant tuberculosis in pregnant patients should be individualized; evidence to support a specific regimen is not available (ATS/CDC/ERS/IDSA [Nahid 2019]; WHO 2020). When second-line agents are needed for the treatment of multidrug-resistant tuberculosis during pregnancy, aminoglycosides should be avoided when alternative agents are effective (ATS/CDC/ERS/IDSA [Nahid 2019]). Amikacin may be considered when an aminoglycoside is needed (HHS [OI adult 2020]).

Amikacin 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. Amikacin is an alternative aminoglycoside recommended for use (in combination with a fluoroquinolone) for treating pregnant patients with bubonic, pharyngeal, pneumonic, or septicemic plague (CDC [Nelson 2021]).

Breastfeeding Considerations

Amikacin is present in breast milk (Matsuda 1984).

Following a single IM dose of amikacin 100 mg given on days 5 to 7 postpartum, only trace amounts of amikacin were seen in breast milk after 4 and 6 hours. Amikacin was not detectable at 1 and 2 hours postdose (Matsuda 1984). In general, antibiotics that are present in breast milk may cause nondose-related modification of bowel flora. Monitor infants for GI disturbances, such as thrush and diarrhea (WHO 2002).

Due to the potential for serious adverse reactions in the breastfeeding infant, the manufacturer recommends a decision be made whether to discontinue breastfeeding or to discontinue the drug, considering the importance of treatment to the mother.

Aminoglycosides have poor oral bioavailability and therefore, use may be considered in breastfeeding patients following treatment for cystic fibrosis (Panchaud 2016).

Patients with multidrug-resistant tuberculosis and a sputum smear-positive test should avoid breastfeeding when possible (Esmail 2018).

Dietary Considerations

Some products may contain sodium.

Monitoring Parameters

Urinalysis, urine output, BUN, serum creatinine, plasma amikacin concentrations (as appropriate to dosing method), vital signs, temperature, weight; test hearing before, during and after treatment, particularly in those at risk for ototoxicity or who will be receiving prolonged therapy (>2 weeks).

Some penicillin derivatives may accelerate the degradation of aminoglycosides in vitro. This may be clinically-significant for certain penicillin (ticarcillin, piperacillin, carbenicillin) and aminoglycoside (gentamicin, tobramycin) combination therapy in patients with significant renal impairment. Close monitoring of aminoglycoside levels is warranted.

Reference Range

Conventional/traditional dosing:

Timing of serum samples: Draw peak 30 minutes after 30-minute infusion has been completed or 1 hour following initiation of infusion or IM injection; draw trough within 30 minutes before next dose. Obtain drug levels after the third dose unless renal dysfunction/toxicity suspected.

Target concentrations: Peak: 20 to 40 mg/L; trough: <8 mg/L (ideal target 1 to 4 mg/L) (Bertino 1994; Drew 2023).

High-dose extended-interval (once-daily) dosing: Obtain a random amikacin level between 6 and 14 hours after the start of the amikacin infusion. Refer to institution-specific nomogram/policies to determine appropriate dosing interval. If an amikacin-specific nomogram is not available, the amikacin level should be divided by 2 and the result evaluated using a gentamicin or tobramycin dosing nomogram. Alternatively, obtain 2 random amikacin levels with the first level ≥1 to 2 hours after the end of infusion and the second level 6 to 12 hours later (≥1 to 2 half-lives apart); adjust dose and interval to achieve goal concentrations below. When therapy is continued for 5 days or more, monitor the amikacin level once or twice weekly (Bailey 1997; Mueller 2009; Nicolau 1995; Prescott 2010).

Target concentrations:

Peak: ~40 mg/L (Leggett 2015; Nicolau 1995); some experts recommend a target peak of 60 mg/L may be needed in critically ill patients (Galvez 2011) or 80 to 120 mg/L in treatment of acute pulmonary exacerbations of cystic fibrosis (Young 2013).

Trough: <2 mg/L (Nicolau 1995).

Intermittent (thrice-weekly) dosing for mycobacterial infection: Target peak: 65 to 80 mcg/mL; target trough: <5 mcg/mL (ATS/ERS/ESCMID/IDSA [Daley 2020]; Peloquin 2004).

Intrathecal/intraventricular (off-label route): Limited data available (IDSA [Tunkel 2017]): Prior to administration of the next intrathecal/intraventricular dose, withdraw a sample of cerebrospinal fluid (CSF). This trough CSF concentration divided by the amikacin minimum inhibitory concentration for the isolated bacterial pathogen (inhibitory quotient) should exceed 10 to 20.

Mechanism of Action

Inhibits protein synthesis in susceptible bacteria by binding to 30S ribosomal subunits

Pharmacokinetics (Adult Data Unless Noted)

Absorption:

IM: Rapid.

Oral: Poorly absorbed.

Distribution: Primarily into extracellular fluid (highly hydrophilic); poor penetration into the blood-brain barrier even when meninges are inflamed.

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

Neonates and Infants ≤6 months: 0.58 L/kg (range: 0.32 to 0.98 L/kg) (Marik 1991).

Infants ≥6 months: 0.5 L/kg (range: 0.22 to 0.73 L/kg) (Marik 1991).

Children and Adolescents ≤15 years: 0.36 ± 0.08 L/kg (Belfayol 1996).

Adults: 0.25 L/kg (Vozeh 1988).

Relative diffusion of antimicrobial agents from blood into CSF: Good only with inflammation (exceeds usual minimum inhibitory concentrations).

CSF:blood level ratio:

Adults: Normal meninges: <10%; Inflamed meninges: ≤25% (MacDougall 2011).

Infants: Normal meninges: 10% to 20%; Inflamed meninges: up to 50%.

Lung:

Bronchial secretions Cmax (peak): serum Cmax (peak) ratio: ~30% to 40%, varies with time (Santré 1995).

Epithelial lining fluid Cmax (peak): Cmax (peak) ratio: ~10% to 18%, likely varies with time (Heffernan 2019; Najmeddin 2020).

Protein binding: 0% to 11%.

Half-life elimination (renal function and age dependent):

Neonates (Howard 1975):

Birth weight <2 kg:

PNA 1 to 3 days: 7.1 hours.

PNA 4 to 7 days: 6.1 hours.

PNA >7 days: 5.5 hours.

Birth weight ≥2 kg:

PNA 1 to 4 days: 6.5 hours.

PNA 4 to 7 days: 5.1 hours.

PNA >7 days: 4.9 hours.

Infants <6 months: 5.02 hours (range: 1.46 to 11.89 hours) (Marik 1991).

Infants ≥6 months: 2.86 hours (range: 0.63 to 6.28 hours) (Marik 1991).

Children and Adolescents ≤15 years: 2 ± 1 hours (Belfayol 1996).

Adults: Normal renal function: ~2 hours; Anuria/end-stage renal disease: 17 to 150 hours (Aronoff 2007).

Time to peak, serum: IM: 60 minutes; IV: Within 30 minutes following a 30-minute infusion; Note: Distribution is prolonged after larger doses (≥60 minutes after 30-minute infusion of 20 mg/kg [Tod 1998]; ≥90 minutes after 60-minute infusion of a high-dose aminoglycoside [gentamicin 7 mg/kg] [Demczar 1997]).

Excretion: Urine (94% to 98% unchanged).

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/minimum inhibitory concentration (MIC), goal: ≥8 to 10 (Kato 2017; Moore 1987; Ruiz 2018; White 2015) or AUC24/MIC, goal: 30 to 50 (mild/moderate infection) or 80 to 100 (severe infection) (Bland 2018; Craig 2011; Drusano 2007; Noel 2019; White 2015).

Expected drug exposure in patients with normal renal function:

Cmax (peak), postdistributional:

Infants and Children:

15 mg/kg: ~24 mg/L (Yu 2015).

7.5 mg/kg: ~15 to 20 mg/L (Alqahtani 2018; Kafetzis 1979).

Adults: 15 mg/kg: ~43 mg/L (Maller 1993).

AUC24: 15 mg/kg: 110 to 145 mg•hour/L (Craig 2011).

Parameters associated with toxicity: Nephrotoxicity is associated with more frequent administration and elevated Cmin (trough) concentrations leading to renal accumulation (Rybak 1999); AUC >300 mg/L has also been found to be predictive (Ruiz 2018).

Postantibiotic effect: Bacterial killing continues after amikacin concentration drops below the MIC of targeted pathogen; generally 0.5 to 7.5 hours, though the actual time of postantibiotic effect varies based on multiple factors including organism, amikacin Cmax (peak), and concomitant antimicrobial therapy (Craig 2011; Lacy 1998; Renneberg 1989).

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

  • (AE) United Arab Emirates: Miacin | Mikacin;
  • (AR) Argentina: Amikacina richet;
  • (AT) Austria: Amikacin b. braun;
  • (AU) Australia: Amikacin | Amikacin wockhardt | Amikin;
  • (BD) Bangladesh: Amimax | Amistar;
  • (BE) Belgium: Amikacine B.Braun | Amikacine hospira | Amikacine mayne pharma (ben);
  • (BG) Bulgaria: Amikin;
  • (BR) Brazil: Amicacil | Amicacina | Amicalin | Amicilon | Amikin | Aminocina | Bactomicin | Klebicil | Sulfato de Amicacina;
  • (CH) Switzerland: Amikin;
  • (CL) Chile: Amikacina;
  • (CN) China: Amikacin | Amikacin sulfate and sodium chloride | Mi ying jie;
  • (CO) Colombia: Amikacina | Amikin | Mikacil;
  • (CZ) Czech Republic: Amikacin b. braun | Amikacin fresenius kabi | Amikacin olikla;
  • (DE) Germany: Amikacin b. braun | Amikacin eberth | Amikacin kabi | Biklin;
  • (DO) Dominican Republic: Amikacina | Furoxin | Pierami;
  • (EC) Ecuador: Amikin;
  • (EG) Egypt: Amikacin | Amikaskiv | Amikin | Emijectacin | Likacin;
  • (ES) Spain: Amikacina combino | Amikacina Mayne | Amikacina normon | Biclin | Kanbine;
  • (FI) Finland: Amikacin macure;
  • (FR) France: Amikacine B.Braun | Amiklin;
  • (GB) United Kingdom: Amikacin;
  • (GR) Greece: Amikacin | Biorisan | Briklin | Farcyclin | Kancin | Lanomycin | Lifermycin | Remikin;
  • (HK) Hong Kong: Amikacin sulphate | Amikin;
  • (HU) Hungary: Amikacin anfarm | Amikacin b. braun | Amikacin kabi | Amikin;
  • (ID) Indonesia: Amikacin | Amikin | Amiosin | Glybotic | Mikaject | Mikasin | Verdix;
  • (IN) India: Aaika | Akasyn | Akik | Alfakim | Alinfec | Amcin | Amee | Amek | Amexel | Amica | Amicid | Amicin | Amicip | Amicom | Amiject | Amikacin | Amikacin sulphate | Amikafine | Amikamac | Amikamed | Amikanex | Amikanit | Amikef | Amimed | Aminat | Amine | Amineg | Aminocin | Amiron | Amisafe | Amistar | Amitax | Amitop | Amity | Amivir | Amk | Amtop | Anamika | Avikacin | Biocin | Bristokacin | Cachmik | Cadicin | Encin | Endocin | Erkacin | Erocin | Eymix | G Kacin | Galmika | Glycacin | Glykacin | Ivimicin | Kacizone | Kami | La mika | Lupamik | M cin | Macmika | Maxee | Mbkacin | Megamica | Megamika | Mica | Mik | Mikabest | Mikacin | Mikadil | Mikaject | Mikarom | Mikastar | Mikatax | Minakin | Moximica | Nimcin | Nosomik | Novacin | Omac | Orkacin | Primikacin | Sermika | Spaike | Tarsocin | Trakacin | Venvicin | Zekacin | Zycin;
  • (IT) Italy: Amikacina bbu | Amikan | Likacin | Mediamik | Migracin | Mikan | Mikavir | Pierami;
  • (JO) Jordan: Amicine | Selemycin;
  • (JP) Japan: Belmaton a;
  • (KE) Kenya: Amicine | Amika | Amikanex | Makcin | Mica;
  • (KR) Korea, Republic of: Aju amikacin | Akicin | Amikacin | Amikacin pm | Amikin | Amiktam | Binex amikacin sulfate | Jw amikacin sulfate premix | Macarin | Myungin amikacin | Samik amikacin | Scd Amikacin | Union amikacin;
  • (KW) Kuwait: Amikacin sulphate;
  • (LB) Lebanon: Amicine;
  • (LT) Lithuania: Amikin | Chemacin;
  • (LV) Latvia: Amikacin | Amikin;
  • (MX) Mexico: Agnicin | Amikason s | Amikin | Antakin | Biclin | Kana;
  • (MY) Malaysia: Amicin | Amikacin | Amikacin DBL | Amikin;
  • (NG) Nigeria: Amcin | Sefkin;
  • (NL) Netherlands: Amikacine;
  • (NO) Norway: Amikacin | Amikacin braun | Amikacin macure | Amikan macure;
  • (NZ) New Zealand: Amikacin | Amikin;
  • (PE) Peru: Amikacina | Amikamek | Amikin | Glumikin;
  • (PH) Philippines: Amikacin Pacific | Quamycin;
  • (PK) Pakistan: Amacin | Amak | Amc 100 injection | Amc 500 injection | Amicin | Amika | Amikaneu | Amikaye | Amikin | Amisave | Amiwise | Elikacin | Glykin | Grasil | Kamerone | Kovex | Medka | Micalpha | Mifate | Mikaton | Romika | Sefkin | Zomacin;
  • (PL) Poland: Amikacin | Amikacin adamed | Amikacin braun | Amikacin kabi | Amikacina normon | Amikin | Biodacyna;
  • (PR) Puerto Rico: Amikin;
  • (PT) Portugal: Amicacina b. braun | Amicacina labesfal | Amikacina normon | Kamina;
  • (PY) Paraguay: Amikacina pasteur | Amikacina sanderson;
  • (QA) Qatar: Amicine | Amikin | Miacin | Mikacin;
  • (RO) Romania: Amikozit | Orlobin;
  • (RU) Russian Federation: Amikin | Selemycin;
  • (SA) Saudi Arabia: Amikacin;
  • (SE) Sweden: Amikacin macure | Biklin;
  • (SG) Singapore: Amikacin | Amikin | Likacin;
  • (SI) Slovenia: Amicasil | Amikacin kabi | Kamina | Selemycin;
  • (SK) Slovakia: Amikacin braun;
  • (TH) Thailand: Akacin | Amackacin | Amicakin | Amicin | Amika | Amikacin | Amikacin injection meiji | Amikasol | Amikin | Amikoside | Pierami | Siamik | Tipkin | Tybikin;
  • (TW) Taiwan: Acemycin | Ami P | Amikacin | Amikin | Amimycin | Aminfec | Eukacin | Orikacin | Pierami | Savox | Sikacin | Unikin;
  • (UA) Ukraine: Amikacid | Amikacin | Amikacin vista | Amikin;
  • (UG) Uganda: Amicip;
  • (VE) Venezuela, Bolivarian Republic of: Amikacina | Amikavax | Behkacin | Biklin;
  • (ZA) South Africa: Amikacin pharma-q | Amikacin sulphate | Amikin | Mikamax
  1. Abbott L, Plummer A, Hoo ZH, Wildman M. Duration of intravenous antibiotic therapy in people with cystic fibrosis. Cochrane Database Syst Rev. 2019;9(9):CD006682. doi:10.1002/14651858.CD006682.pub6 [PubMed 31487382]
  2. Abdel-Hady E, El Hamamsy M, Hedaya M, Awad H. The efficacy and toxicity of two dosing-regimens of amikacin in neonates with sepsis. J Clin Pharm Ther. 2011;36(1):45-52. doi:10.1111/j.1365-2710.2009.01152.x [PubMed 21198719]
  3. Agence française de sécurité sanitaire des produits de santé (ANSM). Update on good use of injectable aminoglycosides, gentamycin, tobramycin, netilmycin, amikacin. Pharmacological properties, indications, dosage, and mode of administration, treatment monitoring. Med Mal Infect. 2012;42(7):301-308. doi:10.1016/j.medmal.2011.07.007 [PubMed 22770656]
  4. Alhadab AA, Ahmed MA, Brundage RC. Amikacin pharmacokinetic-pharmacodynamic analysis in pediatric cancer patients. Antimicrob Agents Chemother. 2018;62(4):e01781-17. doi:10.1128/AAC.01781-17 [PubMed 29358293]
  5. Allegaert K, Anderson BJ, Cossey V, et al. Limited Predictability of Amikacin Clearance in Extreme Premature Neonates at Birth. Br J Clin Pharmacol. 2006;61(1):39-48. [PubMed 16390350]
  6. Allegaert K, Scheers I, Cossey V, et al. Covariates of Amikacin Clearance in Neonates: The Impact of Postnatal Age on Predictability. Drug Metab Lett. 2008;2(4):286-289. [PubMed 19356107]
  7. Alqahtani S, Abouelkheir M, Alsultan A, et al. Optimizing amikacin dosage in pediatrics based on population pharmacokinetic/pharmacodynamic modeling. Paediatr Drugs. 2018;20(3):265-272. doi:10.1007/s40272-018-0288-y [PubMed 29569124]
  8. 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.
  9. American Thoracic Society and Infectious Diseases Society of America. Guidelines for the Management of Adults With Hospital-Acquired, Ventilator-Associated, and Healthcare-Associated Pneumonia. Am J Respir Crit Care Med. 2005;171(4):388-416. [PubMed 15699079]
  10. Amikacin injection [prescribing information]. Schaumburg, IL: Sagent Pharmaceuticals; September 2022.
  11. Antibiotic therapy for children who are obese. In: Bradley JS, Nelson JD, Barnett ED, et al, eds. Nelson's Pediatric Microbial Therapy. 27th ed. American Academy of Pediatrics; 2021:chap 13.
  12. Aréchiga-Alvarado NA, Medellín-Garibay SE, Milán-Segovia RDC, Ortiz-Álvarez A, Magaña-Aquino M, Romano-Moreno S. Population pharmacokinetics of amikacin administered once daily in patients with different renal functions. Antimicrob Agents Chemother. 2020;64(5):e02178-19. doi:10.1128/AAC.02178-19 [PubMed 32041715]
  13. Ariffin H, Arasu A, Mahfuzah M, Ariffin WA, Chan LL, Lin HP. Single-daily ceftriaxone plus amikacin versus thrice-daily ceftazidime plus amikacin as empirical treatment of febrile neutropenia in children with cancer. J Paediatr Child Health. 2001;37(1):38-43. doi:10.1046/j.1440-1754.2001.00585.x [PubMed 11168867]
  14. Armstrong DK, Hodgman T, Visconti JA , Reilley TE, Garner WL, Dasta JF. Hemodialysis of amikacin in critically ill patients. Crit Care Med. 1988;16(5):517-520. doi:10.1097/00003246-198805000-00010 [PubMed 3359790]
  15. 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.
  16. Baddour L, 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 March 10, 2021.
  17. Bailey TC, Little JR, Littenberg B, Rechley RM, Dunagan WC. A meta-analysis of extended interval dosing versus multiple daily dosing of aminoglycosides. Clin Infect Dis. 1997;24(5):786-795. doi:10.1093/clinids/24.5.786 [PubMed 9142771]
  18. 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. [PubMed 26373317]
  19. Bauer LA, Blouin RA. Influence of age on amikacin pharmacokinetics in patients without renal disease. Comparison with gentamicin and tobramycin. Eur J Clin Pharmacol. 1983;24(5):639-642. doi:10.1007/BF00542214 [PubMed 6873143]
  20. Begg EJ, Barclay ML. Aminoglycosides - 50 Years On. Br J Clin Pharmacol. 1995;39(6):597-603. [PubMed 7654476]
  21. Begg EJ, Barclay ML, Kirkpatrick CJ. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharmacol. 1999;47(1):23-30. [PubMed 10073735]
  22. Belfayol L, Talon P, Eveillard M, Alet P, Fauvelle F. Pharmacokinetics of once-daily amikacin in pediatric patients. Clin Microbiol Infect. 1996;2(3):186-191. doi:10.1016/s1198-743x(14)65141-7 [PubMed 11866842]
  23. Bensaid B, Rozieres A, Nosbaum A, Nicolas JF, Berard F. Amikacin-induced drug reaction with eosinophilia and systemic symptoms syndrome: delayed skin test and ELISPOT assay results allow the identification of the culprit drug. J Allergy Clin Immunol. 2012;130(6):1413-1414. doi:10.1016/j.jaci.2012.05.042 [PubMed 22840850]
  24. Bernard B, Abate M, Thielen PF, et al. Maternal-Fetal Pharmacological Activity of Amikacin. J Infect Dis. 1977;135(6):925-932. [PubMed 864287]
  25. Bertino JS Jr, Rodvold KA, Destache CJ. Cost considerations in therapeutic drug monitoring of aminoglycosides. Clin Pharmacokinet. 1994;26(1):71-81. doi:10.2165/00003088-199426010-00006 [PubMed 8137600]
  26. 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]
  27. 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]
  28. 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]
  29. Caceres Guido P, Perez M, Halac A, et al. Population pharmacokinetics of amikacin in patients with pediatric cystic fibrosis. Pediatr Pulmonol. 2019;54(11):1801-1810. doi:10.1002/ppul.24468 [PubMed 31402602]
  30. Canis F, Husson MO, Turck D, et al. Pharmacokinetics and bronchial diffusion of single daily dose amikacin in cystic fibrosis patients. J Antimicrob Chemother. 1997;39(3):431-433. doi:10.1093/jac/39.3.431 [PubMed 9096197]
  31. Centers for Disease Control and Prevention (CDC). Nocardiosis information for healthcare workers. Updated March 30, 2016. Accessed September 2, 2020. https://www.cdc.gov/nocardiosis/health-care-workers/.
  32. Charnas R, Lüthi AR, Ruch W. Once daily ceftriaxone plus amikacin vs. three times daily ceftazidime plus amikacin for treatment of febrile neutropenic children with cancer. Writing Committee for the International Collaboration on Antimicrobial Treatment of Febrile Neutropenia in Children. Pediatr Infect Dis J. 1997;16(4):346-353. doi:10.1097/00006454-199704000-00003 [PubMed 9109134]
  33. Chow MS, Quintiliani R, Nightingale CH. In vivo inactivation of tobramycin by ticarcillin. A case report. JAMA. 1982;247(5):658-659. [PubMed 6798229]
  34. 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]
  35. 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]
  36. 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]
  37. Corpus KA, Weber KB, Zimmerman CR. Intrathecal amikacin for the treatment of pseudomonal meningitis. Ann Pharmacother. 2004;38(6):992-995. doi: 10.1345/aph.1D541. [PubMed 15122005]
  38. Craig WA. Optimizing aminoglycoside use. Crit Care Clin. 2011;27(1):107-121. doi:10.1016/j.ccc.2010.11.006 [PubMed 21144989]
  39. Cristea S, Smits A, Kulo A, et al. Amikacin pharmacokinetics to optimize dosing in neonates with perinatal asphyxia treated with hypothermia. Antimicrob Agents Chemother. 2017;61(12):e01282-17. doi:10.1128/AAC.01282-17 [PubMed 28993332]
  40. Cunha BA, “Aminoglycosides: Current Role in Antimicrobial Therapy,” Pharmacotherapy, 1988, 8(6):334-50. [PubMed 3146747]
  41. Daley CL, Iaccarino JM, Lange C, et al. Treatment of nontuberculous mycobacterial pulmonary disease: an official ATS/ERS/ESCMID/IDSA clinical practice guideline. Clin Infect Dis. 2020;71(4):905-913. doi:10.1093/cid/ciaa1125 [PubMed 32797222]
  42. 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]
  43. Dowell JA, Korth-Bradley J, Milisci M, et al. Evaluating Possible Pharmacokinetic Interactions Between Tobramycin, Piperacillin, and a Combination of Piperacillin and Tazobactam in Patients With Various Degrees of Renal Impairment. J Clin Pharmacol. 2001;41:979-986. [PubMed 11549103]
  44. Drew RH. Dosing and administration of parenteral aminoglycosides. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 8, 2023.
  45. 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]
  46. Duszynska W, Taccone FS, Hurkacz M, Kowalska-Krochmal B, Wiela-Hojeńska A, Kübler A. Therapeutic drug monitoring of amikacin in septic patients. Crit Care. 2013;17(4):R165. doi:10.1186/cc12844 [PubMed 23886243]
  47. Edson RS, Terrell CL. The Aminoglycosides. Mayo Clin Proc. 1999;74(5):519-528. [PubMed 10319086]
  48. Engle WA and American Academy of Pediatrics Committee on Fetus and Newborn. Age Terminology During the Perinatal Period. Pediatrics. 2004;114(5):1362-1364. [PubMed 15520122]
  49. Esmail A, Sabur NF, Okpechi I, Dheda K. Management of drug-resistant tuberculosis in special sub-populations including those with HIV co-infection, pregnancy, diabetes, organ-specific dysfunction, and in the critically ill. J Thorac Dis. 2018;10(5):3102-3118. doi:10.21037/jtd.2018.05.11 [PubMed 29997980]
  50. 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]
  51. 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.
  52. Farchione LA. Inactivation of Aminoglycosides by Penicillins. J Antimicrob Chemother. 1982;8(suppl A):27-36.
  53. Flandrois JP, Bouletreau P, Auboyer R, et al. Accidental Amikacin Overdose in Man: Emergency Therapy by Extrarenal Dialysis. Infection. 1979;7:190-191. [PubMed 511337]
  54. Floto RA, Olivier KN, Saiman L, et al. US Cystic Fibrosis Foundation and European Cystic Fibrosis Society consensus recommendations for the management of non-tuberculous mycobacteria in individuals with cystic fibrosis. Thorax. 2016;71(suppl 1):i1-22. [PubMed 26666259]
  55. Flume PA, Mogayzel PJ Jr, Robinson KA, et al; Clinical Practice Guidelines for Pulmonary Therapies Committee. Cystic fibrosis pulmonary guidelines: treatment of pulmonary exacerbations. Am J Respir Crit Care Med. 2009;180(9):802-808. doi:10.1164/rccm.200812-1845PP [PubMed 19729669]
  56. Fuchs PC, Stickel S, Anderson PH, et al. In Vitro Inactivation of Aminoglycosides by Sulbactam, Other Beta-Lactams, and Sulbactam-Beta-Lactam Combinations. Antimicrob Agents Chemother. 1991;35(1):182-184. [PubMed 2014975]
  57. Gálvez R, Luengo C, Cornejo R, et al Higher than recommended amikacin loading doses achieve pharmacokinetic targets without associated toxicity. Int J Antimicrob Agents. 2011;38(2):146-151. doi:10.1016/j.ijantimicag.2011.03.022 [PubMed 21612894]
  58. Gilbert DN. Once-Daily Aminoglycoside Therapy. Antimicrob Agents Chemother. 1991;35(3):399-405. [PubMed 2039189]
  59. Gilbert VE, Beals JD, Natelson ST, et al. Treatment of Cerebrospinal Fluid Leaks and Gram-negative Bacillary Meningitis With Large Doses of Intrathecal Amikacin and Systemic Antibiotics. Neurosurgery. 1986;18(4):402-406. [PubMed 3754626]
  60. Gilbert DN, Leggett JE. Aminoglycosides. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 7th ed. Churchill Linvingstone Elsevier; 2009.
  61. Green FJ, Lavelle KJ, Aronoff GR. Management of Amikacin Overdose. Am J Kidney Dis. 1981;1:110-112. [PubMed 7332003]
  62. Griffith DE, Aksamit T, Brown-Elliott BA, et al. An Official ATS/IDSA Statement: Diagnosis, Treatment, and Prevention of Nontuberculous Mycobacterial Diseases. Am J Respir Crit Care Med. 2007;175(4):367-416. doi:10.1164/rccm.200604-571ST [PubMed 17277290]
  63. Guadalupe Vásquez-Mendoza M, Vargas-Origel A, Del Carmen Ramos-Jiménez A, Aguilar-Orozco G, Romero-Gutiérrez G. Efficacy and renal toxicity of one daily dose of amikacin versus conventional dosage regime. Am J Perinatol. 2007;24(2):141-146. doi:10.1055/s-2006-958159 [PubMed 17304422]
  64. Guardado AR, Blanco A, Asensi V, et al. Multidrug-Resistant Acinetobacter Meningitis in Neurosurgical Patients With Intraventricular Catheters: Assessment of Different Treatments. J Antimicrob Chemother. 2008;61(4):908-913. [PubMed 18281693]
  65. Halstenson CE, Wong MO, Herman CS, et al. Effect of Concomitant Administration of Piperacillin on the Dispositions on Isepamicin and Gentamicin in Patients With End-Stage Renal Disease. Antimicrob Agents Chemother. 1992;36(9):1832-1836. [PubMed 1416875]
  66. Hashimoto Y, Shima T, Matsukawa S, Satou M. A possible hazard of prolonged neuromuscular blockade by amikacin. Anesthesiology. 1978;49(3):219-220. doi:10.1097/00000542-197809000-00018 [PubMed 686450]
  67. 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]
  68. Hassan NA, Awdallah FF, Abbassi MM, Sabry NA. Nebulized versus IV amikacin as adjunctive antibiotic for hospital and ventilator-acquired pneumonia postcardiac surgeries: a randomized controlled trial. Crit Care Med. 2018;46(1):45-52. doi:10.1097/CCM.0000000000002695 [PubMed 28857848]
  69. Haworth CS, Banks J, Capstick T, et al. British Thoracic Society guidelines for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). Thorax. 2017;72(suppl 2):ii1-ii64. doi:10.1136/thoraxjnl-2017-210927 [PubMed 29054853]
  70. 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]
  71. 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):562-77. [PubMed 19397464]
  72. Hitt CM, Patel KB, Nicolau DP, et al. Influence of Piperacillin-Tazobactam on Pharmacokinetics of Gentamicin Given Once Daily. Am J Health Syst Pharm. 1997;54(23):2704-2708. [PubMed 9408514]
  73. Ho PW, Pien FD, Kominami N. Massive Amikacin Overdose. Ann Intern Med. 1979;91:227-228. [PubMed 464467]
  74. Howard JB, McCracken GH. Pharmacological evaluation of Amikacin in Neonates. Antimicrob Agents Chemother. 1975; 8(1):86-90. [PubMed 1164009]
  75. Hughes KM, Johnson PN, Anderson MP, Sekar KC, Welliver RC, Miller JL. Comparison of amikacin pharmacokinetics in neonates following implementation of a new dosage protocol. J Pediatr Pharmacol Ther. 2017;22(1):33-40. doi:10.5863/1551-6776-22.1.33 [PubMed 28337079]
  76. Illamola SM, Colom H, van Hasselt JG. Evaluating renal function and age as predictors of amikacin clearance in neonates: model-based analysis and optimal dosing strategies. Br J Clin Pharmacol. 2016;82(3):793-805. doi:10.1111/bcp.13016 [PubMed 27198625]
  77. Iseman MD. Treatment of Multidrug-Resistant Tuberculosis. N Engl J Med. 1993;329(11):784-791. [PubMed 8350889]
  78. Jenh AM, Tamma PD, Milstone AM. Extended-interval aminoglycoside dosing in pediatrics. Pediatr Infect Dis J. 2011;30(4):338-339. [PubMed 21407038]
  79. Kafetzis DA, Sinaniotis CA, Papadatos CJ, Kosmidis J. Pharmacokinetics of amikacin in infants and pre-school children. Acta Paediatr Scand. 1979;68(3):419-422. doi:10.1111/j.1651-2227.1979.tb05030.x [PubMed 443042]
  80. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):e61-e111. doi:10.1093/cid/ciw353 [PubMed 27418577]
  81. Kasiakou SK, Rafailidis PI, Liaropoulos K, et al. Cure of Post-Traumatic Recurrent Multiresistant Gram-Negative Rod Meningitis With Intraventricular Colistin. J Infect. 2005; 50(4):348-352. [PubMed 15845435]
  82. Kato H, Hagihara M, Hirai J, et al. Evaluation of amikacin pharmacokinetics and pharmacodynamics for optimal initial dosing regimen. Drugs R D. 2017;17(1):177-187. doi:10.1007/s40268-016-0165-5 [PubMed 28063020]
  83. Kenyon CF, Knoppert DC, Lee SK, et al. Amikacin Pharmacokinetics and Suggested Dosage Modifications for the Preterm Infant. Antimicrob Agents Chemother. 1990;34(2):265-268. [PubMed 2327775]
  84. Khan SA, Waqas M, Siddiqui UT, et al. Intrathecal and intraventricular antibiotics for postoperative gram-negative meningitis and ventriculitis. Surg Neurol Int. 2017;8:226. doi:10.4103/sni.sni_81_17 [PubMed 29026662]
  85. Klompas M. Treatment of hospital-acquired and ventilator-associated pneumonia in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 20, 2022.
  86. 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]
  87. Konishi H, Goto M, Nakamoto Y, et al. Tobramycin Inactivation by Carbenicillin, Ticarcillin, and Piperacillin. Antimicrob Agents Chemother. 1983;23(5):653-657. [PubMed 6223576]
  88. 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. doi:10.1023/a:1015968407226 [PubMed 2762224]
  89. Labaune JM, Bleyzac N, Maire P, et al. Once-a-Day Individualized Amikacin Dosing for Suspected Infection at Birth Based on Population Pharmacokinetic Models. Biol Neonate. 2001;80(2):142-147. [PubMed 11509814]
  90. 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]
  91. Lafont E, Conan PL, Rodriguez-Nava V, Lebeaux D. Invasive nocardiosis: disease presentation, diagnosis and treatment - old questions, new answers? Infect Drug Resist. 2020;13:4601-4613. doi:10.2147/IDR.S249761 [PubMed 33376366]
  92. Lau A, Lee M, Flascha S, et al. Effect of Piperacillin on Tobramycin Pharmacokinetics in Patients With Normal Renal Function. Antimicrob Agents Chemother. 1983;24(4):533-537. [PubMed 6651279]
  93. Lau WK, Young LS, Osher AB, Dooley RR. Amikacin therapy of exacerbations of Pseudomonas aeruginosa infections in patients with cystic fibrosis. Pediatrics. 1977;60(3):372-377. [PubMed 408787]
  94. Le J, Ashley ED, Neuhauser MM, et al. Consensus summary of aerosolized antimicrobial agents: application of guideline criteria. Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2010;30(6):562-584. [PubMed 20500046]
  95. Leggett JE. Aminoglycosides. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Elsevier Saunders; 2015. [PubMed Leggett.1]
  96. Leroy A, Humbert G, Oksenhendler G, Fillastre JP. Comparative pharmacokinetics of lividomycin, amikacin and sisomicin in normal subjects and in uraemic patients. J Antimicrob Chemother. 1976;2(4):373-381. doi:10.1093/jac/2.4.373 [PubMed 1052072]
  97. 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]
  98. Li PKT, Szeto CC, Piraino B, et al. Peritoneal Dialysis-Related Infections Recommendations: 2010 Update. Peritoneal Dialysis International. 2010;30:393-423. [PubMed 20628102]
  99. Liu X, Smits A, Wang Y, et al. Impact of disease on amikacin pharmacokinetics and dosing in children. Ther Drug Monit. 2019;41(1):44-52. doi:10.1097/FTD.0000000000000568 [PubMed 30299427]
  100. Lortholary O, Tod M, Cohen Y, et al. Aminoglycosides. Med Clin North Am. 1995;79(4):761-787. [PubMed 7791422]
  101. MacDougall C, Chambers HF, eds. Goodman & Gilman's: The Pharmacological Basis of Therapeutics. 12th ed. McGraw-Hill Global Education Holding; 2011.
  102. Mahmoudi L, Mohammadpour AH, Ahmadi A, Niknam R, Mojtahedzadeh M. Influence of sepsis on higher daily dose of amikacin pharmacokinetics in critically ill patients. Eur Rev Med Pharmacol Sci. 2013;17(3):285-291. [PubMed 23426530]
  103. Maller R, Ahrne H, Holmen C, Lausen I, Nilsson LE, Smedjegård J; Scandinavian Amikacin Once Daily Study Group. Once- versus twice-daily amikacin regimen: efficacy and safety in systemic gram-negative infections. J Antimicrob Chemother. 1993;31(6):939-948. doi:10.1093/jac/31.6.939 [PubMed 8360131]
  104. Marik PE, Havlik I, Monteagudo FS, Lipman J. The pharmacokinetic of amikacin in critically ill adult and paediatric patients: comparison of once- versus twice-daily dosing regimens. J Antimicrob Chemother. 1991;27(Suppl C):S81-S89. doi:10.1093/jac/27.suppl_c.81 [PubMed 1856148]
  105. Matsuda S. Transfer of Antibiotics Into Maternal Milk. Biol Res Pregnancy Perinatol. 1984;5(2):57-60. [PubMed 6743732]
  106. 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. doi:10.1089/sur.2016.261 [PubMed 28085573]
  107. McCormack JP, Jewesson PJ. A Critical Re-evaluation of the “Therapeutic Range” of Aminoglycosides. Clin Infect Dis. 1992;14(1):320-339.
  108. 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]
  109. Medjebeur Hanna R, Levy M, Bille E, et al. Assessment of the effects of a high amikacin dose on plasma peak concentration in critically ill children. Paediatr Drugs. 2021;23(4):395-401. doi:10.1007/s40272-021-00456-0 [PubMed 34142330]
  110. Miele K, Bamrah Morris S, Tepper NK. Tuberculosis in pregnancy. Obstet Gynecol. 2020;135(6):1444-1453. doi:10.1097/AOG.0000000000003890 [PubMed 32459437]
  111. 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. doi:10.2146/ajhp140363 [PubMed 25465576]
  112. Moffett BS, Morris J, Galati M, Munoz F, Arikan AA. Population pharmacokinetics of vancomycin in pediatric extracorporeal membrane oxygenation. Pediatr Crit Care Med. 2018;19(10):973-980. [PubMed 30063652]
  113. 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]
  114. 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]
  115. 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.
  116. Nahid P, Dorman SE, Alipanah N, et al. Official American Thoracic Society/Centers for Disease Control and Prevention/Infectious Diseases Society of America Clinical Practice Guidelines: treatment of drug-susceptible tuberculosis. Clin Infect Dis. 2016;63(7):e147-e195. [PubMed 27516382]
  117. Nahid P, Mase SR, Migliori GB, et al. Treatment of drug-resistant tuberculosis. An official ATS/CDC/ERS/IDSA clinical practice guideline. Am J Respir Crit Care Med. 2019;200(10):e93-e142. doi:10.1164/rccm.201909-1874ST [PubMed 31729908]
  118. Najmeddin F, Shahrami B, Azadbakht S, et al. Evaluation of epithelial lining fluid concentration of amikacin in critically ill patients with ventilator-associated pneumonia. J Intensive Care Med. 2020;35(4):400-404. doi:10.1177/0885066618754784 [PubMed 29471721]
  119. 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]
  120. Nemecek BD, Hammond DA. Demystifying drug dosing in renal dysfunction. American Society of Health-System Pharmacists; 2019.
  121. 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]
  122. Nicholson TT, Smith A, McKone EF, Gallagher CG. Duration of intravenous antibiotic treatment for acute exacerbations of cystic fibrosis: A systematic review: Duration of treatment for acute exacerbations of cystic fibrosis. J Cyst Fibros. Published online September 27, 2021. doi:10.1016/j.jcf.2021.08.017 [PubMed 34588142]
  123. Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW, Quintiliani R. Experience with a once-daily aminoglycoside program administered to 2184 adult patients. Antimicrob Agents Chemother. 1995;39(3):650-655. doi:10.1128/aac.39.3.650 [PubMed 7793867]
  124. Niederman MS, Alder J, Bassetti M, et al. Inhaled amikacin adjunctive to intravenous standard-of-care antibiotics in mechanically ventilated patients with gram-negative pneumonia (INHALE): a double-blind, randomised, placebo-controlled, phase 3, superiority trial. Lancet Infect Dis. 2020;20(3):330-340. doi:10.1016/S1473-3099(19)30574-2 [PubMed 31866328]
  125. Noel AR, Attwood M, Bowker KE, Kim A, Krause KM, MacGowan AP. Pharmacodynamics of plazomicin and a comparator aminoglycoside, amikacin, studied in an in vitro pharmacokinetic model of infection. Int J Antimicrob Agents. 2019;54(5):626-632. doi:10.1016/j.ijantimicag.2019.07.001 [PubMed 31299297]
  126. 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]
  127. Ott SR, Meier N, Kolditz M, et al; OPINION study group. Pulmonary nocardiosis in Western Europe-Clinical evaluation of 43 patients and population-based estimates of hospitalization rates. Int J Infect Dis. 2019;81:140-148. doi:10.1016/j.ijid.2018.12.010 [PubMed 30658169]
  128. 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]
  129. 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]
  130. Panchaud A, Di Paolo ER, Koutsokera A, et al. Safety of drugs during pregnancy and breastfeeding in cystic fibrosis patients. Respiration. 2016;91(4):333-348. [PubMed 26942733]
  131. Peloquin CA, Berning SE, Nitta AT, et al. Aminoglycoside toxicity: daily versus thrice-weekly dosing for treatment of mycobacterial diseases. Clin Infect Dis. 2004;38(11):1538-1544. doi:10.1086/420742 [PubMed 15156439]
  132. Prescott WA Jr. National survey of extended-interval aminoglycoside dosing in pediatric cystic fibrosis pulmonary exacerbations. J Pediatr Pharmacol Ther. 2011;16(4):262-269. doi:10.5863/1551-6776-16.4.262 [PubMed 22768010]
  133. 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]
  134. Preston SL, Briceland LL. Intrathecal administration of amikacin for treatment of meningitis secondary to cephalosporin-resistant Escherichia coli. Ann Pharmacother. 1993;27(7-8):870-873. doi:10.1177/106002809302700709 [PubMed 8364266]
  135. Preston SL, Briceland LL. Single Daily Dosing of Aminoglycosides. Pharmacotherapy. 1995;15(3):297-316. [PubMed 7667165]
  136. Public Health Service Task Force on Prophylaxis and Therapy for Mycobacterium avium Complex. Recommendations on Prophylaxis and Therapy for Disseminated Mycobacterium avium Complex Disease in Patients Infected With the Human Immunodeficiency Virus. N Engl J Med. 1993;329(12):898-904. [PubMed 8395019]
  137. Qin JP, Huang HB, Zhou H, Zhu Y, Xu Y, Du B. Amikacin nebulization for the adjunctive therapy of gram-negative pneumonia in mechanically ventilated patients: a systematic review and meta-analysis of randomized controlled trials. Sci Rep. 2021;11(1):6969. doi:10.1038/s41598-021-86342-8 [PubMed 33772055]
  138. Refer to manufacturer's labeling.
  139. Renneberg J, Walder M. Postantibiotic effects of imipenem, norfloxacin, and amikacin in vitro and in vivo. Antimicrob Agents Chemother. 1989;33(10):1714-1720. doi:10.1128/aac.33.10.1714 [PubMed 2511798]
  140. Restrepo A, Clark NM; Infectious Diseases Community of Practice of the American Society of Transplantation. Nocardia infections in solid organ transplantation: guidelines from the Infectious Diseases Community of Practice of the American Society of Transplantation. Clin Transplant. 2019;33(9):e13509. doi:10.1111/ctr.13509 [PubMed 30817024]
  141. Results of the Endophthalmitis Vitrectomy Study. A randomized trial of immediate vitrectomy and of intravenous antibiotics for the treatment of postoperative bacterial endophthalmitis. Endophthalmitis Vitrectomy Study Group. Arch Ophthalmol. 1995;113(12):1479-1496. [PubMed 7487614]
  142. Roger C, Wallis SC, Muller L, et al. Influence of renal replacement modalities on amikacin population pharmacokinetics in critically ill patients on continuous renal replacement therapy. Antimicrob Agents Chemother. 2016;60(8):4901-4909. doi:10.1128/AAC.00828-16 [PubMed 27270279]
  143. Roth DB, Flynn HW Jr. Antibiotic Selection in the Treatment of Endophthalmitis: The Significance of Drug Combinations and Synergy. Surv Ophthalmol. 1997;41(5):395-401. [PubMed 9163836]
  144. 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]
  145. Ruiz J, Ramirez P, Company MJ, et al. Impact of amikacin pharmacokinetic/pharmacodynamic index on treatment response in critically ill patients. J Glob Antimicrob Resist. 2018;12:90-95. doi:10.1016/j.jgar.2017.09.019 [PubMed 29017888]
  146. Russoe ME, Atkins-Thor E. Gentamicin and Ticarcillin in Subjects With End-Stage Renal Disease. Comparison of Two Assay Methods and Evaluation of Inactivation Rate. Clin Nephrol. 1981;15(4):175-180. [PubMed 7237864]
  147. 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]
  148. 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]
  149. Santré C, Georges H, Jacquier JM, et al. Amikacin levels in bronchial secretions of 10 pneumonia patients with respiratory support treated once daily versus twice daily. Antimicrob Agents Chemother. 1995;39(1):264-267. doi:10.1128/aac.39.1.264 [PubMed 7695320]
  150. Schaad UB, Wedgwood-Krucko J, Guenin K, Buehlmann U, Kraemer R. Antipseudomonal therapy in cystic fibrosis: aztreonam and amikacin versus ceftazidime and amikacin administered intravenously followed by oral ciprofloxacin. Eur J Clin Microbiol Infect Dis. 1989;8(10):858-865. doi:10.1007/BF01963771 [PubMed 2512129]
  151. Schaad UB, Wedgwood-Krucko J, Suter S, Kraemer R. Efficacy of inhaled amikacin as adjunct to intravenous combination therapy (ceftazidime and amikacin) in cystic fibrosis. J Pediatr. 1987;111(4):599-605. [PubMed 3309236]
  152. Sherwin CM, Svahn S, Van der Linden A, et al. Individualised Dosing of Amikacin in Neonates: A Pharmacokinetic/Pharmacodynamic Analysis. Eur J Clin Pharmacol. 2009;65(7):705-713. [PubMed 19305985]
  153. Shofty B, Neuberger A, Naffaa ME, et al. Intrathecal or intraventricular therapy for post-neurosurgical gram-negative meningitis: matched cohort study. Clin Microbiol Infect. 2016;22(1):66-70. doi:10.1016/j.cmi.2015.09.023 [PubMed 26456474]
  154. Smetana KS, Cook AM. Cerebrospinal fluid vancomycin dosing and monitoring: the quandary posed by guideline recommendations. Clin Infect Dis. 2018;67(6):980-981. doi:10.1093/cid/ciy189 [PubMed 29518176]
  155. Smits A, De Cock RF, Allegaert K, Vanhaesebrouck S, Danhof M, Knibbe CA. Prospective evaluation of a model-based dosing regimen for amikacin in preterm and term neonates in clinical practice. Antimicrob Agents Chemother. 2015;59(10):6344-6351. doi:10.1128/AAC.01157-15 [PubMed 26248375]
  156. Solomkin JS, Mazuski JE, Bradley JS, et al. Diagnosis and management of complicated intra-abdominal infections in adults and children: guidelines by the Surgical Infection Society and the Infectious Diseases Society of America. Clin Infect Dis. 2010;50(2):133-164. [PubMed 20034345]
  157. 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.
  158. Taccone FS, de Backer D, Laterre PF, et al. Pharmacokinetics of a loading dose of amikacin in septic patients undergoing continuous renal replacement therapy. Int J Antimicrob Agents. 2011;37(6):531-535. doi:10.1016/j.ijantimicag.2011.01.026 [PubMed 21489756]
  159. Thompson MIB, Russo ME, Saxon BJ, et al. Gentamicin Inactivation by Piperacillin or Carbenicillin in Patients With End-Stage Renal Disease. Antimicrob Agents Chemother. 1982;21(2):268-273. [PubMed 6462107]
  160. Tod M, Lortholary O, Seytre D, et al. Population pharmacokinetic study of amikacin administered once or twice daily to febrile, severely neutropenic adults. Antimicrob Agents Chemother. 1998;42(4):849-856. [PubMed 9559795]
  161. Touw DJ, Westerman EM, Sprij AJ. Therapeutic drug monitoring of aminoglycosides in neonates. Clin Pharmacokinet. 2009;48(2):71-88. [PubMed 19271781]
  162. 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. [PubMed 7726530]
  163. 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):1159-1166. [PubMed 16163635]
  164. Tréluyer JM, Merlé Y, Tonnelier S, Rey E, Pons G. Nonparametric population pharmacokinetic analysis of amikacin in neonates, infants, and children. Antimicrob Agents Chemother. 2002;46(5):1381-1387. doi:10.1128/AAC.46.5.1381-1387.2002 [PubMed 11959572]
  165. 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]
  166. 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. doi:10.1093/cid/ciw861 [PubMed 28203777]
  167. 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]
  168. Urban AW, Craig WA. Daily dosage of aminoglycosides. Curr Clin Top Infect Dis. 1997;17:236-255. [PubMed 9189668]
  169. US Department of Health and Human Services (HHS) Panel on Opportunistic Infections in Adults and Adolescents with HIV. Guidelines for 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. http://aidsinfo.nih.gov/contentfiles/lvguidelines/adult_oi.pdf. Accessed April 27, 2020.
  170. 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/en/guidelines/adult-and-adolescent-opportunistic-infection/whats-new-guidelines. Updated November 18, 2021. Accessed February 1, 2022.
  171. US Department of Health and Human Services (HHS) Panel on Opportunistic Infections in HIV-Exposed and HIV-Infected Children. Guidelines for prevention and treatment of opportunistic infections in HIV-exposed and HIV-infected children. https://clinicalinfo.hiv.gov/en/guidelines/pediatric-opportunistic-infection/whats-new. Updated March 19, 2021. Accessed February 1, 2022.
  172. Van der Auwera P. Pharmacokinetic Evaluation of Single Daily Dose Amikacin. J Antimicrob Chemother. 1991;27(suppl C):63-71.
  173. VanDevanter DR, Flume PA, Morris N, Konstan MW. Probability of IV antibiotic retreatment within thirty days is associated with duration and location of IV antibiotic treatment for pulmonary exacerbation in cystic fibrosis. J Cyst Fibros. 2016;15(6):783-790. doi:10.1016/j.jcf.2016.04.005 [PubMed 27139161]
  174. Vanhaeverbeek M, Siska G, Douchamps J, Herchuelz A. Comparison of the Efficacy and Safety of Amikacin Once or Twice-a-Day in the Treatment of Severe Gram-Negative Infections in the Elderly. Int J Clin Pharmacol Ther Toxicol. 1993;31(3):153-156. [PubMed 8468114]
  175. Vic P, Ategbo S, Turck D, et al. Tolerance, pharmacokinetics and efficacy of once daily amikacin for treatment of Pseudomonas aeruginosa pulmonary exacerbations in cystic fibrosis patients. Eur J Pediatr. 1996;155(11):948-953. doi:10.1007/BF02282885 [PubMed 8911895]
  176. Viollier AF, Standiford HC, Drusano GL, et al, "Comparative Pharmacokinetics and Serum Bactericidal Activity of Mezlocillin, Ticarcillin and Piperacillin, With and Without Gentamicin," J Antimicrob Chemother, 1985, 15(5):597-606. [PubMed 4008387]
  177. Vozeh S, Schmidlin O, Taeschner W. Pharmacokinetic Drug Data. Clin Pharmacokinetics. 1988;15(4):254-282. [PubMed 3191648]
  178. Wallace CS, Hall M, Kuhn RJ. Pharmacologic management of cystic fibrosis. Clin Pharm. 1993;12(9):657-674. [PubMed 8306566]
  179. Walterspiel JN, Feldman S, Van R, et al. Comparative Inactivation of Isepamicin, Amikacin, and Gentamicin by Nine Beta-Lactams and Two Beta-Lactamase Inhibitors, Cilastatin and Heparin. Antimicrob Agents Chemother. 1991;35(9):1875-1878. [PubMed 1952861]
  180. Wang JH, Lin PC, Chou CH, et al. Intraventricular antimicrobial therapy in postneurosurgical gram-negative bacillary meningitis or ventriculitis: a hospital-based retrospective study. J Microbiol Immunol Infect. 2014;47(3):204-210. doi:10.1016/j.jmii.2012.08.028 [PubMed 23201321]
  181. 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-S86. [PubMed 22851742]
  182. White BP, Lomaestro B, Pai MP. Optimizing the initial amikacin dosage in adults. Antimicrob Agents Chemother. 2015;59(11):7094-7096. doi:10.1128/AAC.01032-15 [PubMed 26282426]
  183. Wilson JW. Nocardiosis: updates and clinical overview. Mayo Clin Proc. 2012;87(4):403-407. doi:10.1016/j.mayocp.2011.11.016 [PubMed 22469352]
  184. 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):1736-1754. doi:10.1161/CIRCULATIONAHA.106.183095 [PubMed 17446442]
  185. Wirt TC, McGee ZA, Oldfield EH, Meacham WF. Intraventricular administration of amikacin for complicated Gram-negative meningitis and ventriculitis. J Neurosurg. 1979;50(1):95-99. doi:10.3171/jns.1979.50.1.0095 [PubMed 363982]
  186. Woillard JB, Bouchet S, Fayon M, Marquet P, Monchaud C, Bui S. A population pharmacokinetic modeling approach to determine the efficacy of intravenous amikacin in children with cystic fibrosis. Ther Drug Monit. 2021;43(4):499-504. doi:10.1097/FTD.0000000000000855 [PubMed 33346630]
  187. World Health Organization (WHO). WHO Treatment Guidelines for Drug-Resistant Tuberculosis, 2016 Update. Geneva: World Health Organization; 2016. http://www.who.int/tb/areas-of-work/drug-resistant-tb/treatment/resources/en/
  188. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the eleventh WHO model list of essential drugs. 2002. http://www.who.int/maternal_child_adolescent/documents/55732/en/
  189. World Health Organization (WHO). WHO consolidated guidelines on tuberculosis. Module 4: treatment - drug-resistant tuberculosis treatment. https://www.who.int/publications/i/item/9789240007048. Published 2020.
  190. Yasuhara H, Kobayashi S, Sakamoto K, et al. Pharmacokinetics of Amikacin and Cephalothin in Bedridden Elderly Patients. J Clin Pharmacol. 1982;22(8-9):403-409. [PubMed 7130429]
  191. Young DC, Zobell JT, Stockmann C, et al. Optimization of anti-pseudomonal antibiotics for cystic fibrosis pulmonary exacerbations: V. Aminoglycosides. Pediatr Pulmonol. 2013;48(11):1047-1061. doi:10.1002/ppul.22813 [PubMed 24000183]
  192. Yu T, Stockmann C, Healy DP, et al. Determination of optimal amikacin dosing regimens for pediatric patients with burn wound sepsis. J Burn Care Res. 2015;36(4):e244-e252. doi:10.1097/BCR.0000000000000159 [PubMed 25185930]
  193. 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 119165 Version 141.0

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