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

Streptomycin: Drug information

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

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

The risk of severe neurotoxic reactions is sharply increased in patients with impaired renal function or prerenal azotemia. These include disturbances of vestibular and cochlear function, optic nerve dysfunction, peripheral neuritis, arachnoiditis, and encephalopathy. The incidence of clinically detectable, irreversible vestibular damage is particularly high in patients treated with streptomycin.

Renal function should be monitored carefully; patients with renal impairment and/or nitrogen retention should receive reduced doses. The peak serum concentration in individuals with kidney damage should not exceed 20 to 25 mcg/mL.

The concurrent or sequential use of other neurotoxic and/or nephrotoxic drugs with streptomycin, including neomycin, kanamycin, gentamicin, cephaloridine, paromomycin, viomycin, polymyxin B, colistin, tobramycin, and cyclosporine should be avoided.

The neurotoxicity of streptomycin can result in respiratory paralysis from neuromuscular blockage, especially when the drug is given soon after the use of anesthesia or muscle relaxants.

The administration of streptomycin in parenteral form should be reserved for patients where adequate laboratory and audiometric testing facilities are available during therapy.

Brand Names: Canada
  • Streptomycin
Pharmacologic Category
  • Antibiotic, Aminoglycoside;
  • Antitubercular Agent
Dosing: Adult

Note: Route of administration: Manufacturer’s labeling states for IM administration only; however, IV administration (off-label route) has been described (Morris 1994; Peloquin 1992; Tanoira 2014).

Aminoglycoside dosing weight: For patients who are underweight (ie, total body weight [TBW] < ideal body weight [IBW]), calculate the dose based on TBW. For patients without obesity (ie, TBW 1 to 1.25 × IBW), calculate the dose based on TBW or IBW. TBW may be preferred in patients without obesity who may have increased volume of distribution (eg, critically ill). For patients with obesity (ie, TBW > 1.25 × IBW), calculate the dose based on adjusted body weight (IBW + [0.4 × (TBW-IBW)]) (Blackburn 2015; Drew 2020). Therapeutic drug monitoring: Monitoring of serum concentrations is recommended to ensure efficacy and avoid toxicity; confirm availability of rapid streptomycin serum concentrations. Timing and frequency of concentration monitoring is individualized based on dosing and monitoring strategy (AHA [Baddour 2015]; Drew 2020; Park 2015; Zhu 2001).

Usual dosage range: IM, IV: 15 to 30 mg/kg/day or 1 to 2 g daily.

Indication-specific dosing:

Brucellosis

Brucellosis: IM, IV: 1 g once daily in combination with doxycycline. Duration depends on extent of disease; streptomycin is usually given for the first 14 to 21 days of therapy, followed by doxycycline monotherapy (Ariza 2007; Bosilkovski 2020; Hasanjani Roushan 2006; Skalsky 2008). Note: Additional agents or other regimens are preferred for neurobrucellosis, endocarditis, and infection in pregnant women (Bosilkovski 2020).

Endocarditis

Endocarditis (alternate agent):

Enterococcus spp. (native or prosthetic valve, susceptible to penicillin and streptomycin/resistant to gentamicin): IM, IV: 7.5 mg/kg every 12 hours in combination with ampicillin or penicillin; Duration of therapy: 4 weeks (native valve and symptoms present <3 months); ≥6 weeks (native valve and symptoms present ≥3 months or prosthetic valve). Note: For native valve endocarditis due to ampicillin-susceptible E. faecalis, a combination regimen that does not contain streptomycin is preferred (AHA [Baddour 2015]).

Mycobacterial infection

Mycobacterial (nontuberculous) (M. avium complex) infection (off-label use):

Pulmonary disease (severe nodular/bronchiectatic or cavitary disease) (adjunctive agent): IM, IV: 10 to 15 mg/kg once daily or 15 to 25 mg/kg 3 times weekly (ATS/ERS/ESCMID/IDSA [Daley 2020]; Peloquin 2004) or 10 to 15 mg/kg 3 times weekly (Kasperbauer 2020) or 15 mg/kg once daily (usual maximum dose: 1 g) for 1 month then 15 mg/kg 3 times weekly (BTS [Haworth 2017]). Give streptomycin as part of an appropriate combination regimen for the first 2 to 3 months of therapy (ATS/ERS/ESCMID/IDSA [Daley 2020]). Note: For older adults or patients who require >6 months of parenteral therapy, some experts recommend empiric dose reduction (BTS [Haworth 2017]).

Pulmonary disease (cavitary or severe bronchiectatic or acid-fast bacilli smear-positive respiratory samples) or systemic illness in patients with cystic fibrosis (adjunctive agent): IM, IV: 15 mg/kg once daily (maximum: 1 g daily) as part of an appropriate combination regimen (CFF/ECFS [Floto 2016]).

Disseminated disease in patients with HIV with CD4 count <50 cells/mm3, high mycobacterial loads (ie, >2 log colony-forming units/mL of blood), or in the absence of effective antiretroviral therapy (adjunctive agent): IM, IV: 1 g once daily as part of an appropriate combination regimen (HHS [OI adult 2019]).

Plague, treatment

Plague (Yersinia pestis), treatment:

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

IM, IV: 1 g every 12 hours for 7 to 14 days and for at least a few days after clinical resolution (CDC [Nelson 2021]; Stout 2022).

Tuberculosis

Tuberculosis (alternative agent): IM, IV: 15 mg/kg once daily or 25 mg/kg 3 times weekly (ATS/CDC/ERS/IDSA [Nahid 2019]; ATS/CDC/IDSA [Nahid 2016]).

Tularemia

Tularemia (alternative agent): IM: 1 g twice daily for ≥10 days depending on severity (CDC 2019; WHO 2007).

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

There are no dosage adjustments provided in the manufacturer’s labeling; however, the following adjustments have been recommended:

Aronoff 2007: Note: Recommendations are based on doses of 1 to 2 g every 6 to 12 hours (1 g once daily for tuberculosis):

CrCl >50 mL/minute: No dosage adjustment necessary.

CrCl 10 to 50 mL/minute: Administer every 24 to 72 hours.

CrCl <10 mL/minute: Administer every 72 to 96 hours.

End-stage renal disease (ESRD):

Intermittent hemodialysis (IHD): One-half the recommended dose administered after hemodialysis on dialysis days. Note: Dosing dependent on the assumption of 3 times weekly complete IHD sessions.

Peritoneal dialysis (PD): Administration via PD fluid: 20 to 40 mg/L (20 to 40 mcg/mL) of PD fluid.

CRRT: Administer every 24 to 72 hours; monitor levels. Note: Drug clearance is highly dependent on the method of renal replacement, filter type, and flow rate. Appropriate dosing requires close monitoring of pharmacologic response, signs of adverse reactions due to drug accumulation, as well as drug concentrations in relation to target trough (if appropriate).

ATS/CDC/IDSA [Nahid 2016]:

Tuberculosis:

CrCl ≥30 mL/minute: No dosage adjustment necessary.

CrCl <30 mL/minute: 15 mg/kg/dose 2 to 3 times weekly.

ESRD on IHD: 15 mg/kg/dose 2 to 3 times weekly. Give after dialysis if given on day of dialysis.

Dosing: Hepatic Impairment: Adult

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

Dosing: Older Adult

Dose reductions are recommended by the manufacturer in patients >60 years of age.

M. avium complex disease: IM/IV: 8 to 10 mg/kg 2 to 3 times weekly (maximum dose for >50 years of age: 500 mg) (ATS/IDSA [Griffith 2007]) or 10 mg/kg once daily for the first month followed by 15 mg/kg 3 times weekly (maximum dose for >59 years of age: 750 mg) (BTS [Haworth 2017]).

Dosing: Pediatric

(For additional information see "Streptomycin: Pediatric drug information")

Note: Initial dosing recommendations presented. Monitoring of serum concentrations is recommended to ensure efficacy and avoid toxicity.

Dosing consideration for obesity: In pediatric patients with obesity, 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 (Koshida 1989; Traynor 1995). Alternatively, adjusted body weight for aminoglycoside dosing may be calculated for patients who are obese using the equation 0.7 × TBW (Bradley 2022). 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 (Moffett 2018). Dosage should then be individualized based on serum concentration monitoring.

General dosing: Infants, Children, and Adolescents: IM, IV: 20 to 40 mg/kg/day in divided doses every 12 to 24 hours; maximum dose: 1,000 mg/dose (Bradley 2022; manufacturer's labeling).

Endocarditis, enterococcal

Endocarditis, enterococcal (alternative agent): Note: Though not included in current pediatric endocarditis guidelines, streptomycin has been recommended for consideration in enterococcal endocarditis for gentamicin-resistant isolates (AHA [Baddour 2015]; AHA [Baltimore 2015]; Bradley 2022).

Infants, Children, and Adolescents: IM, IV: 20 to 30 mg/kg/day in divided doses every 12 hours as part of an appropriate combination regimen; maximum dose: 1,000 mg/dose (AHA [Baddour 2005]; manufacturer's labeling). Duration should be at least 4 to 6 weeks depending on whether prosthetic material is involved and which concomitant agent is used (AHA [Baltimore 2015]).

Mycobacterial infection, nontuberculous

Mycobacterial infection, nontuberculous:

Buruli ulcers (Mycobacterium ulcerans infection): Limited data available: Infants, Children, and Adolescents: IM, IV: 15 mg/kg once daily; maximum dose: 1,000 mg/dose. Use as part of an appropriate combination regimen for 8 weeks (CDC 2015; WHO 2012).

Disseminated mycobacterium avium complex disease in patients with HIV (adjunctive agent): Note: May be used as the third or fourth drug in patients with CD4 count <50 cells/mm3, high mycobacterial loads (ie, >2 log colony-forming units/mL of blood), or in the absence of effective antiretroviral therapy (HHS [OI adult] 2023).

Adolescents: IM, IV: 1,000 mg once daily as part of an appropriate combination regimen (HHS [OI adult] 2023).

Pulmonary infection in patients with cystic fibrosis: Infants, Children, and Adolescents: IM, IV: 20 to 40 mg/kg/dose once daily as part of an appropriate combination regimen; maximum dose: 1,000 mg/dose. Overall treatment duration is ≥12 months after culture conversion. Note: When streptomycin is used, it is part of the initial, intensive phase of treatment; continuation phase may not include streptomycin depending on culture results (CFF/ECFS [Floto 2016]).

Pulmonary infection in patients without cystic fibrosis: Infants, Children, and Adolescents: IM: 15 mg/kg/dose once daily as part of an appropriate combination regimen; after initial period, may decrease to 15 mg/kg/dose 3 times weekly; maximum dose: 1,000 mg/dose. Overall duration is ≥12 months after culture conversion (BTS [Haworth 2017]).

Plague

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

Infants, Children, and Adolescents: IM, IV: 30 mg/kg/day divided every 12 hours for 10 to 14 days; maximum dose: 1,000 mg/dose (CDC [Nelson 2021]).

Tuberculosis disease [active tuberculosis]; drug-resistant

Tuberculosis disease (active tuberculosis); drug-resistant (alternative agent): Note: Recommended only in patients in whom susceptibility is confirmed and when absolutely necessary (ie, a 5-drug regimen of more effective or less toxic medications is not possible); always 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 (ATS/CDC/ERS/IDSA [Nahid 2019]; Red Book [AAP 2021]; WHO 2022).

Once-daily regimen:

Infants, Children, and Adolescents: IM, IV: 15 to 20 mg/kg/dose once daily; some experts recommend doses of 20 to 40 mg/kg/dose once daily; maximum dose: 1,000 mg/dose (ATS/CDC/ERS/IDSA [Nahid 2019]; ATS/CDC/IDSA [Nahid 2016]; Red Book [AAP 2021]; WHO 2022).

Twice-weekly regimen: Note: Once-daily therapy is typically preferred. Dosing modified from adult dosing and accounting for increased total body water (ATS/CDC/ERS/IDSA [Nahid 2019]).

Infants, Children, and Adolescents: IM, IV: 25 to 30 mg/kg/dose twice weekly (ATS/CDC/ERS/IDSA [Nahid 2019]; ATS/CDC/IDSA [Nahid 2016]).

Tuberculosis disease [active tuberculosis]; meningitis

Tuberculosis disease (active tuberculosis); meningitis (alternative agent): Note: Shorter, more intensive regimens including ethionamide may be preferred (Solomons 2022; WHO 2022).

Infants, Children, and Adolescents: IM, IV: 20 to 40 mg/kg/dose once daily; doses as low as 15 mg/kg/dose once daily have been recommended; maximum daily dose: 1,000 mg/day. Streptomycin may be discontinued once isolate is determined susceptible to first-line drugs (ATS/CDC/IDSA [Nahid 2016]; HHS [OI pediatric] 2022; Red Book [AAP 2021]).

Tularemia

Tularemia (alternative agent): Infants, Children, and Adolescents: IM: 30 to 40 mg/kg/day in divided doses every 12 hours for 10 days; maximum dose: 1,000 mg/dose. In mild illness, 5 to 7 days of therapy may be sufficient; longer courses may be necessary for severe disease (Red Book [AAP 2021]; WHO 2007).

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

There are no adjustments provided in the manufacturer's labeling; however, the following adjustments have been recommended (Aronoff 2007):

Infants, Children, and Adolescents: Note: Renally adjusted dose recommendations are based on doses of 20 to 40 mg/kg/day every 24 hours. Monitor serum concentrations.

GFR 30 to 50 mL/minute/1.73 m2: Administer 7.5 mg/kg/dose every 24 hours.

GFR 10 to 29 mL/minute/1.73 m2: Administer 7.5 mg/kg/dose every 48 hours.

GFR <10 mL/minute/1.73 m2: Administer 7.5 mg/kg/dose every 72 to 96 hours.

Intermittent hemodialysis (IHD): Administer 7.5 mg/kg/dose every 72 to 96 hours.

Peritoneal dialysis (PD): Administer 7.5 mg/kg/dose every 72 to 96 hours.

Continuous renal replacement therapy (CRRT): Administer every 24 hours; monitor levels. Note: Drug clearance is highly dependent on the method of renal replacement, filter type, and flow rate. Appropriate dosing requires close monitoring of pharmacologic response, signs of adverse reactions due to drug accumulation, as well as drug concentrations in relation to target trough (if appropriate).

Dosing: Hepatic Impairment: Pediatric

There are no 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.

Cardiovascular: Hypotension

Central nervous system: Drug fever, facial paresthesia, headache, neurotoxicity

Dermatologic: Exfoliative dermatitis, skin rash, urticaria

Gastrointestinal: Nausea, vomiting

Genitourinary: Azotemia, nephrotoxicity

Hematologic & oncologic: Eosinophilia, hemolytic anemia, leukopenia, pancytopenia, thrombocytopenia

Hypersensitivity: Anaphylaxis, angioedema

Neuromuscular & skeletal: Arthralgia, tremor, weakness

Ophthalmic: Amblyopia

Otic: Auditory ototoxicity, vestibular ototoxicity

Respiratory: Dyspnea

<1%, postmarketing, and/or case reports: DRESS syndrome (drug reaction with eosinophilia and systemic symptoms), toxic epidermal necrolysis

Contraindications

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

Warnings/Precautions

Concerns related to adverse effects:

• 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, including disturbances of vestibular and cochlear function, optic nerve dysfunction, peripheral neuritis, arachnoiditis, and encephalopathy; usual risk factors include pre-existing renal impairment, concomitant neuro-/nephrotoxic medications. 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. Baseline and periodic caloric stimulation and audiometric tests are recommended with prolonged therapy. 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 pre-existing vertigo, tinnitus, or hearing loss.

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

• Renal impairment: [US Boxed Warning]: May cause nephrotoxicity. Use with caution in patients with renal impairment; dose adjustment necessary in patients with renal impairment and/or nitrogen retention. Monitor renal function closely; peak serum concentrations should not surpass 20 to 25 mcg/mL in patients with renal impairment.

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 with other neurotoxic and/or nephrotoxic drugs (eg, neomycin, kanamycin, gentamicin, paromomycin, polymyxin B, colistin, tobramycin, cyclosporine).

Dosage form specific issues:

• Sulfite sensitivity: Some formulations may contain sodium metabisulfite; may cause allergic reactions including anaphylaxis or asthma exacerbations (some life-threatening) in susceptible patients.

Other warnings/precautions:

• Appropriate use: [US Boxed Warning]: Parenteral form should be used only where appropriate audiometric and laboratory testing facilities are available. IM injections should be administered in a large muscle well within the body to avoid peripheral nerve damage and local skin reactions.

Dosage Forms: US

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

Solution Reconstituted, Intramuscular [preservative free]:

Generic: 1 g (1 ea)

Generic Equivalent Available: US

Yes

Pricing: US

Solution (reconstituted) (Streptomycin Sulfate Intramuscular)

1 g (per each): $93.75

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 Reconstituted, Intramuscular:

Generic: 1 g (5 mL)

Administration: Adult

IM: Inject deep IM into large muscle mass; midlateral thigh muscle or upper outer quadrant of buttocks; rotate injection sites.

IV (off-label route): After dilution in admixture, infuse over 30 to 60 minutes (Morris 1994; Peloquin 1992; Tanoira 2014)

Administration: Pediatric

Parenteral:

IM: Inject deep IM into a large muscle mass appropriate for age; rotate injection sites.

IV: After further dilution, infuse over 30 to 60 minutes (Morris 1994; Peloquin 1992; Pérez Tanoira 2014).

Use: Labeled Indications

Tuberculosis:

Treatment of tuberculosis, in combination with other appropriate antituberculosis agents, when the primary agents are contraindicated because of toxicity or intolerance.

Nontuberculosis infections:

Treatment of infections caused by susceptible bacteria that are not amenable to therapy with less potentially toxic agents, including sensitive Yersinia pestis (plague); Francisella tularensis (tularemia); Brucella; Klebsiella granulomatis (donovanosis, granuloma inguinale); Haemophilus ducreyi (chancroid); Haemophilus influenzae (in respiratory, endocardial, and meningeal infections, concomitantly with another antibacterial agent); Klebsiella pneumoniae pneumonia (concomitantly with another antibacterial agent); Escherichia coli, Proteus spp., Klebsiella aerogenes (formerly Enterobacter aerogenes), K. pneumoniae, and Enterococcus faecalis in urinary tract infections; Streptococcus viridans; E. faecalis (in endocardial infections, concomitant with penicillin); and gram-negative bacillary bacteremia (concomitant with another antibacterial agent).

Use: Off-Label: Adult

Mycobacterium avium complex infection

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

Streptomycin may be confused with streptozocin

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): Streptomycin may enhance the nephrotoxic effect of Bacitracin (Systemic). 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

Streptomycin crosses the placenta.

Streptomycin may cause fetal harm if administered to a pregnant patient. There are multiple reports of total irreversible bilateral congenital deafness in children whose mothers received streptomycin during pregnancy.

Streptomycin is approved for the treatment of tuberculosis (TB). 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 TB 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 TB during pregnancy, aminoglycosides should be avoided when alternative agents are effective (ATS/CDC/ERS/IDSA [Nahid 2019]). Streptomycin is not the preferred agent when an aminoglycoside is required for the treatment of TB during pregnancy (HHS [OI adult 2019]).

Untreated plague (Yersinia pestis) infection 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. Streptomycin is an alternative aminoglycoside recommended for use (in combination with a fluroquinolone) for treating pregnant patients with bubonic, pharyngeal, pneumonic, or septicemic plague (CDC [Nelson 2021]).

Breastfeeding Considerations

Streptomycin is present in breast milk.

Aminoglycosides have poor oral bioavailability (Edson 1991), which may limit systemic absorption via breast milk.

Streptomycin is considered compatible with breastfeeding. In general, antibiotics that are present in breast milk may cause non-dose-related modification of bowel flora. Monitor infants for GI disturbances, such as thrush and diarrhea (WHO 2002). Patients with multidrug resistant tuberculosis and a sputum smear-positive test should avoid breastfeeding when possible (Esmail 2018).

Monitoring Parameters

Baseline and periodic hearing tests (audiograms), clinical assessment for vertigo and tinnitus, BUN, creatinine, serum electrolytes; serum drug concentrations should be monitored in all patients (ATS/CDC/ERS/IDSA [Nahid 2019]).

Reference Range

Therapeutic:

Peak: Daily dosing: 20 to 35 mcg/mL (35 to 45 mcg/mL for mycobacterial disease); Three times weekly dosing: 65 to 80 mcg/mL (AHA [Baddour 2015]; ATS/ERS/ESCMID/IDSA [Daley 2020]; BTS [Haworth 2017]; CFF/ECFS [Floto 2016]; Edson 1991; Peloquin 2004; Zhu 2001).

Trough: <5 mcg/mL (AHA [Baddour 2015]; BTS [Haworth 2017]; CFF/ECFS [Floto 2016]; Edson 1991; Zhu 2001).

Supratherapeutic: Trough: >10 mcg/mL.

Mechanism of Action

Inhibits bacterial protein synthesis by binding directly to the 30S ribosomal subunits causing faulty peptide sequence to form in the protein chain

Pharmacokinetics (Adult Data Unless Noted)

Absorption: Oral: Poorly absorbed; IM: Well absorbed.

Distribution: Distributes into most body tissues and fluids except the brain; small amounts enter the cerebrospinal fluid only with inflamed meninges.

Vd:

Neonates (GA ≥33 weeks, PNA 4 to 20 days): 0.24 to 0.44 L/kg (Herngren 1977).

Infants ≥6 months and Children: Mean range: 0.25 ± 0.02 to 0.29 ± 0.03 L/kg (Bolme 1988).

Protein binding: 34%.

Half-life elimination:

Neonates:

Preterm, PNA 1 to 3 days: Mean: 7 hours; range: 4 to 9.1 hours (Axline 1964).

Preterm and term (GA ≥33 weeks), PNA 4 to 20 days: 1.39 to 4.64 hours (Herngren 1977).

Infants ≥6 months and Children: Mean range: 1.69 ± 0.36 to 2.58 ± 0.44 hours (Bolme 1988).

Adults: ~2 to 4.7 hours; prolonged with renal impairment.

Time to peak: IM: Within 1 to 2 hours.

Excretion: Urine (29% to 89% as unchanged drug); small amount (1%) excreted in bile, saliva, sweat, and tears.

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Anti-infective considerations:

Parameters associated with efficacy: Gram-negative bacilli: Concentration dependent; associated with Cmax (peak)/minimum inhibitory concentration (MIC), goal: ≥8 to 10 (Abdul-Aziz 2020; Lacy 1998) or AUC24/MIC, goal: 30 to 50 (mild/moderate infection, patients who are immunocompetent) or 80 to 100 (severe infection) (Bland 2018; Craig 2011; Drusano 2007).

Expected drug exposure in normal renal function:

Cmax (peak): Adults:

IV: 14 mg/kg (range: 11 to 25 mg/kg) (30-minute infusion): 43.6 mg/L (range: 9 to 107 mg/L) (Zhu 2001).

IM: 18 mg/kg (range: 10 to 23 mg/kg): 42.6 mg/L (range: 2.9 to 85.2 mg/L) (Zhu 2001).

IM: 1 g: 25 to 50 mg/L (manufacturer's labeling).

AUC24: Adults:

IV: 14 mg/kg (range: 11 to 25 mg/kg) (30-minute infusion): 284 mg•hour/L (range: 124 to 680 mg•hour/L) (Zhu 2001).

IM: 18 mg/kg (range: 10 to 23 mg/kg): 264 mg•hour/L (range: 175 to 343 mg•hour/L) (Zhu 2001).

Parameters associated with toxicity: Nephrotoxicity is associated with high Cmin (trough) aminoglycoside concentrations leading to renal accumulation (Abdul-Aziz 2020; Bertino 1993; Rybak 1999; Verpooten 1989).

Postantibiotic effect: Bacterial killing continues after streptomycin concentration falls below the MIC of targeted pathogen and varies significantly based on the organism, duration of antimicrobial exposure, streptomycin concentration, and concomitant antimicrobial therapy:

E. faecalis/E. faecium (alone or in combination with ampicillin or vancomycin): ~0 to 2 hours (Fuursted 1987; Fuursted 1989).

Mycobacterium tuberculosis: 32.2 hours (range: 0.7 to 60.2 hours) (Chan 2001).

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

  • (AE) United Arab Emirates: Streptomycin;
  • (AR) Argentina: Estreptomicina | Estreptomicina richet;
  • (AU) Australia: Streptomycin;
  • (BD) Bangladesh: Streptomycin;
  • (BE) Belgium: Streptomycine sulfate pfizer;
  • (BG) Bulgaria: Streptomycin;
  • (BR) Brazil: Furp estreptomicina;
  • (CN) China: Streptomycin;
  • (CO) Colombia: Estreptomicina;
  • (CZ) Czech Republic: Strepto Fatol | Streptomycin;
  • (DE) Germany: Strepto Fatol | Strepto-Hefa | Streptomycin | Streptothenat;
  • (DO) Dominican Republic: Estreptomicina;
  • (EC) Ecuador: Estreptomicina | Estreptomicina Sulfate;
  • (EE) Estonia: Streptomycin | Streptomycin tzf | Streptomycine panpharma | Streptomycini sulfas;
  • (EG) Egypt: Streptomycin;
  • (ES) Spain: Estrepto P H | Estreptomicina normon | Neodualestrepto;
  • (FI) Finland: Streptomycin;
  • (FR) France: Streptomycine panpharm;
  • (GB) United Kingdom: Streptaquaine | Streptomycin;
  • (GR) Greece: Streptomycine;
  • (HK) Hong Kong: Streptomycin | Streptomycin sulphate;
  • (HU) Hungary: Streptomycin | Streptomycin egis;
  • (ID) Indonesia: Streptomycin | Streptomycin sulphate;
  • (IE) Ireland: Streptomycin | Streptomycin sulphate;
  • (IL) Israel: Streptomycin;
  • (IN) India: Ambistryn-s | Cipstryn | Streptobiotic | Streptomac | Streptomycin;
  • (IT) Italy: Streptomicina solfato;
  • (JP) Japan: Streptomycin sulfate daiichi | Streptomycin sulfate kyowa | Streptomycin sulfate nikken;
  • (KE) Kenya: Strepto 1 | Streptomycin | Streptomycin sulphate | Streptomycin sulphate reig jofre;
  • (LB) Lebanon: Streptomycine;
  • (LT) Lithuania: Ambistryn-s | Estreptomicina normon | Streptomycin | Streptomycin sulphate | Streptomycini sulfas;
  • (LV) Latvia: Streptomycin | Streptomycine;
  • (MA) Morocco: Streptomycine specia;
  • (MX) Mexico: Estreptomicina | Estreptomicina s | Sulfestrep | Treptorim;
  • (MY) Malaysia: Streptin | Streptomycin | Streptomycin sulphate;
  • (NG) Nigeria: Everdestiny streptomycin sulphate;
  • (NO) Norway: Strepto Fatol;
  • (PE) Peru: Estreptomicina;
  • (PH) Philippines: Pharmawealth streptomycin | Septin;
  • (PK) Pakistan: Streptomycin;
  • (PL) Poland: Pan streptomycin | Streptomycin;
  • (PT) Portugal: Estreptomicin azev | Estreptomicina | Estreptomicina atral | Estreptomicina labesfal;
  • (RU) Russian Federation: Streptomycin | Streptomycin kmp | Streptomycin sulph;
  • (SA) Saudi Arabia: Streptomycin;
  • (SG) Singapore: Streptomycin sulphate meiji;
  • (SI) Slovenia: Strepto Fatol | Strepto-Hefa;
  • (SK) Slovakia: Streptomycin;
  • (TH) Thailand: Streptomycin | Streptomycin meiji | Streptomycin sulphate;
  • (TN) Tunisia: Streptomycine | Streptomycine panpharma;
  • (TR) Turkey: Strep | Streptomycine;
  • (TW) Taiwan: Streptomycin;
  • (UA) Ukraine: Streptomycin sulfa;
  • (UG) Uganda: Ustrep;
  • (UY) Uruguay: Estreptomicina;
  • (ZA) South Africa: Novostrep | Solustrep | Streptomycin | Streptomycin sulphate;
  • (ZM) Zambia: Streptocin | Streptomycin | Streptomycin sulphate;
  • (ZW) Zimbabwe: Streptomycin sulphate
  1. Abdul-Aziz MH, Alffenaar JC, Bassetti M, et al; Infection Section of European Society of Intensive Care Medicine (ESICM); Pharmacokinetic/pharmacodynamic and Critically Ill Patient Study Groups of European Society of Clinical Microbiology and Infectious Diseases (ESCMID); Infectious Diseases Group of International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT); Infections in the ICU and Sepsis Working Group of International Society of Antimicrobial Chemotherapy (ISAC). Antimicrobial therapeutic drug monitoring in critically ill adult patients: a position paper. Intensive Care Med. 2020;46(6):1127-1153. doi:10.1007/s00134-020-06050-1 [PubMed 32383061]
  2. Abughali N, Van der Kuyp F, Annable W, Kumar ML. Congenital tuberculosis. Pediatr Infect Dis J. 1994;13(8):738-741. doi:10.1097/00006454-199408000-00012 [PubMed 7970976 ]
  3. Ad Hoc Committee of the Scientific Assembly on Microbiology, Tuberculosis, and Pulmonary Infections. Treatment of Tuberculosis and Tuberculosis Infection in Adults and Children. Clin Infect Dis. 1995;21:9-27.
  4. American Academy of Pediatrics (AAP). In: Kimberlin DW, Barnett ED, Lynfield R, Sawyer MH, eds. Red Book: 2021 Report of the Committee on Infectious Diseases. 32nd ed. American Academy of Pediatrics; 2021.
  5. American Thoracic Society (ATS); CDC; Infectious Diseases Society of America. Treatment of tuberculosis. MMWR Recomm Rep. 2003;52(RR-11):1-77. http://www.cdc.gov/mmwr/preview/mmwrhtml/rr5211a1.htm [PubMed 12836625]
  6. Ariza J, Bosilkovski M, Cascio A, et al. Perspectives for the treatment of brucellosis in the 21st century: the Ioannina recommendations. PLoS Med. 2007;4(12):e317. doi:10.1371/journal.pmed.0040317 [PubMed 18162038]
  7. 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.
  8. Axline SG, Simon HJ. Clinical pharmacology of antimicrobials in premature infants. I. Kanamycin, streptomycin, and neomycin. Antimicrob Agents Chemother (Bethesda). 1964;10:135-141. [PubMed 14287917]
  9. Baddour LM, Wilson WR, Bayer AS, et al; American Heart Association Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young, Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and Stroke Council. Infective endocarditis in adults: diagnosis, antimicrobial therapy, and management of complications: a scientific statement for healthcare professionals from the American Heart Association. Circulation. 2015;132(15):1435-1486. doi:10.1161/CIR.0000000000000296 [PubMed 26373316]
  10. Baddour LM, Wilson WR, Bayer AS, et al; Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease in the Young, Councils on Clinical Cardiology, Stroke, and Cardiovascular Surgery and Anesthesia, American Heart Association, Infectious Diseases Society of America. AHA scientific statement: Infective endocarditis: diagnosis, antimicrobial therapy, and management of complications: a statement for healthcare professionals from the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease in the Young, and the Councils on Clinical Cardiology, Stroke, and Cardiovascular Surgery and Anesthesia, American Heart Association: endorsed by the Infectious Diseases Society of America. Circulation. 2005;111(23):e394-434. doi:10.1161/CIRCULATIONAHA.105.165564 [PubMed 15956145]
  11. Baltimore RS, Gewitz M, Baddour LM, et al. Infective endocarditis in childhood: 2015 update: a scientific statement from the American Heart Association (AHA). Circulation. 2015;132(15):1487-1515. [PubMed 26373317]
  12. Balyan FR, Taibah A, De Donato G, et al. Titration streptomycin therapy in Meniere's disease: long-term results. Otolaryngol Head Neck Surg. 1998;118(2):261-266. doi:10.1016/S0194-5998(98)80028-4 [PubMed 9482564]
  13. Begg EJ, Barclay ML. Aminoglycosides - 50 Years On. Br J Clin Pharmacol. 1995;39(6):597-603. [PubMed 7654476]
  14. Bertino JS Jr, Booker LA, Franck PA, Jenkins PL, Franck KR, Nafziger AN. Incidence of and significant risk factors for aminoglycoside-associated nephrotoxicity in patients dosed by using individualized pharmacokinetic monitoring. J Infect Dis. 1993;167(1):173-179. doi:10.1093/infdis/167.1.173 [PubMed 8418164]
  15. 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]
  16. 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]
  17. Bolme P, Eriksson M, Habte D, Paalzow L. Pharmacokinetics of streptomycin in Ethiopian children with tuberculosis and of different nutritional status. Eur J Clin Pharmacol. 1988;33(6):647-649. doi:10.1007/BF00542504 [PubMed 3259181]
  18. Bosilkovski M. Brucellosis: treatment and prevention. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed April 29, 2020.
  19. Bradley JS, Nelson JD, Kimberlin DK, et al, eds. Nelson's Pocket Book of Pediatric Antimicrobial Therapy. 28th ed. American Academy of Pediatrics; 2022.
  20. Campbell IA, Drobinewski F, Novelli V, et al. Management of Opportunistic Mycobacterial Infections: Joint Tuberculosis Committee Guidelines 1999. Subcommittee of the Joint Tuberculosis Committee of the British Thoracic Society. Thorax. 2000;55(3):210-218. doi:10.1136/thorax.55.3.210 [PubMed 10679540]
  21. Cantwell MF, Shehab ZM, Costello AM, et al. Brief report: congenital tuberculosis. N Engl J Med. 1994;330(15):1051-1054. doi:10.1056/NEJM199404143301505 [PubMed 8127333 ]
  22. Centers for Disease Control and Prevention (CDC). Buruli ulcer. https://www.cdc.gov/buruli-ulcer/health-care-workers.html. Updated January 26, 2015. Accessed May 26, 2023.
  23. Centers for Disease Control and Prevention (CDC). Tickborne diseases of the United States. https://www.cdc.gov/ticks/tickbornediseases/tularemia.html. Updated January 10, 2019. Accessed May 12, 2020.
  24. Chan CY, Au-Yeang C, Yew WW, Hui M, Cheng AF. Postantibiotic effects of antituberculosis agents alone and in combination. Antimicrob Agents Chemother. 2001;45(12):3631-3634. doi:10.1128/AAC.45.12.3631-3634.2001 [PubMed 11709357]
  25. Craig WA. Optimizing aminoglycoside use. Crit Care Clin. 2011;27(1):107-121. doi:10.1016/j.ccc.2010.11.006 [PubMed 21144989]
  26. Cunha BA. Aminoglycosides: current role in antimicrobial therapy. Pharmacotherapy. 1988;8(6):334-350. doi:10.1002/j.1875-9114.1988.tb04092.x [PubMed 3146747]
  27. 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]
  28. Davidson PT, Le HQ. Drug treatment of tuberculosis - 1992. Drugs. 1992;43(5):651-673. doi:10.2165/00003495-199243050-00003 [PubMed 1379145]
  29. Drew RH. Dosing and administration of parenteral aminoglycosides. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed April 29, 2020.
  30. Drugs for Tuberculosis. Med Lett Drugs Ther. 1993;35(908):99-101. [PubMed 8412982]
  31. 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]
  32. Edson RS, Terrell CL. The aminoglycosides. Mayo Clin Proc. 1991;66(11):1158-1164. [PubMed 1943249]
  33. 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]
  34. Fleck-Derderian S, Nelson CA, Cooley KM, et al. Plague during pregnancy: a systematic review. Clin Infect Dis. 2020;70(suppl 1):S30-S36. doi:10.1093/cid/ciz1228 [PubMed 32435806]
  35. 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–i22. doi:10.1136/thoraxjnl-2015-207360 [PubMed 26666259]
  36. Fuursted K. Comparison of the post-antibiotic effect of Streptococcus faecalis and Streptococcus faecium with ampicillin alone or combined with streptomycin: studies on a novel type of antimicrobial interaction. Acta Pathol Microbiol Immunol Scand B. 1987;95(6):351-354. doi:10.1111/j.1699-0463.1987.tb03137.x [PubMed 3125717]
  37. Fuursted K. Synergism and mechanism of subinhibitory concentration of streptomycin on Streptococcus faecalis. APMIS. 1989;97(1):27-32. doi:10.1111/j.1699-0463.1989.tb00750.x [PubMed 2492428]
  38. Gould FK, Denning DW, Elliott TS, et al. Guidelines for the Diagnosis and Antibiotic Treatment of Endocarditis in Adults: A Report of the Working Party of the British Society for Antimicrobial Chemotherapy. J Antimicrob Chemother. 2012;67(2):269-289. [PubMed 22086858]
  39. Hasanjani Roushan MR, Mohraz M, Hajiahmadi M, Ramzani A, Valayati AA. Efficacy of gentamicin plus doxycycline versus streptomycin plus doxycycline in the treatment of brucellosis in humans. Clin Infect Dis. 2006;42(8):1075‐1080. doi:10.1086/501359 [PubMed 16575723]
  40. Havlir DV, Barnes PF. Tuberculosis in patients with human immunodeficiency virus infection. N Engl J Med. 1999;340(5):367-373. [PubMed 9929528]
  41. 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]
  42. Herngren L, Boréus LO, Jalling B, Lagercrantz R. Pharmacokinetic aspects of streptomycin treatment of neonatal septicemia. Scand J Infect Dis. 1977;9(4):301-308. doi:10.3109/inf.1977.9.issue-4.09 [PubMed 601524]
  43. Hmouda H, Laouani-Kechrid C, Karoui M, et al. A Rare Case of Streptomycin-Induced Toxic Epidermal Necrolysis in a Patient With Tuberculosis: A Therapeutic Dilemma. Ann Pharmacother. 2005;39(1):165-168. [PubMed 15546942]
  44. Iseman MD. Treatment of Multidrug-Resistant Tuberculosis. N Engl J Med. 1993;329(11):784-791. [PubMed 8350889]
  45. Kasperbauer S, Daley CL. Treatment of Mycobacterium avium complex pulmonary infection in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed December 23, 2020.
  46. Kim-Sing A, Kays MB, Vivien EJ, et al. Intravenous Streptomycin Use in a Patient Infected With High-Level Gentamicin-Resistant Streptococcus faecalis .Ann Pharmacother. 1993;27(6):712-714. [PubMed 8329788]
  47. 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]
  48. 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]
  49. Miele K, Bamrah Morris S, Tepper NK. Tuberculosis in pregnancy. Obstet Gynecol. 2020;135(6):1444-1453. doi:10.1097/AOG.0000000000003890 [PubMed 32459437]
  50. Moffett BS, Kam C, Galati M, et al. The "ideal" body weight for pediatric gentamicin dosing in the era of obesity: a population pharmacokinetic analysis. Ther Drug Monit. 2018;40(3):322-329. doi:10.1097/FTD.0000000000000505 [PubMed 29521784]
  51. Morris JT, Cooper RH. Intravenous streptomycin: a useful route of administration. Clin Infect Dis. 1994;19(6):1150-1151. [PubMed 7888550]
  52. 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. doi: 10.1093/cid/ciw376 [PubMed 27516382]
  53. 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]
  54. Nelson CA, Meaney-Delman D, Fleck-Derderian S, et al; 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]
  55. Newberry DM, Robertson Bell T. Congenital tuberculosis: a new concern in the neonatal intensive care unit. Adv Neonatal Care. 2018;18(5):341-349. doi:10.1097/ANC.0000000000000555 [PubMed 30096058]
  56. Okascharoen C, Nuntnarumit P, Sirinavin S. Neonatal tuberculosis associated with shock, disseminated intravascular coagulation, hemophagocytic syndrome, and hypercalcemia: a case report. J Perinatol. 2003;23(1):79-81. [PubMed 12556935]
  57. Park SI, Oh J, Jang K, et al. Pharmacokinetics of second-line antituberculosis drugs after multiple administrations in healthy volunteers. Antimicrob Agents Chemother. 2015;59(8):4429‐4435. doi:10.1128/AAC.00354-15 [PubMed 25987620]
  58. Passeron T, Ndir M, Aubron C, et al. Drug rash with eosinophilia and systemic symptoms (DRESS) due to streptomycin. Acta Derm Venerol. 2004;84(1):92-93. doi:10.1080/00015550310006095 [PubMed 15040497]
  59. Peloquin CA. Shortages of antimycobacterial drugs. N Engl J Med. 1992;326(10):714. doi:10.1056/NEJM199203053261018 [PubMed 1736124]
  60. Peloquin CA. Therapeutic drug monitoring in the treatment of tuberculosis. Drugs. 2002;62(15):2169-2183. doi:10.2165/00003495-200262150-00001 [PubMed 12381217]
  61. 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]
  62. Pérez Tanoira R, Sánchez-Patán F, Jiménez Girón A, Fernández Roblas R, Esteban J, Fernández Guerrero ML. Tolerance and safety of intravenous streptomycin therapy in patients with tuberculosis. Infection. 2014;42(3):597-598. doi:10.1007/s15010-014-0621-5 [PubMed 24748409]
  63. Prevention and treatment of tuberculosis among patients infected with human immunodeficiency virus: principles of therapy and revised recommendations. Centers for Disease Control and Prevention. MMWR Recomm Rep. 1998;47(RR-20):1-58. [PubMed 9809743]
  64. 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. doi:10.1128/AAC.43.7.1549 [PubMed 10390201]
  65. Schaaf HS, Garcia-Prats AJ, Donald PR. Antituberculosis drugs in children. Clin Pharmacol Ther. 2015;98(3):252-265. doi:10.1002/cpt.164 [PubMed 26095192]
  66. Schütte D, Umboock A, Pluschke G. Phagocytosis of Mycobacterium ulcerans in the Course of Rifampicin and Streptomycin Chemotherapy in Buruli Ulcer Lesions. Br J Derm. 2009;160(2):273-283. doi:10.1111/j.1365-2133.2008.08879.x [PubMed 19016694]
  67. Shao Y, Hageman JR, Shulman ST. Congenital and perinatal tuberculosis. Neoreviews. 2021;22(9):e600-e605. doi:10.1542/neo.22-9-e600 [PubMed 34470761]
  68. Skalsky K, Yahav D, Bishara J, et al. Treatment of Human Brucellosis: Systematic Review and Meta-Analysis of Randomised Controlled Trials. BMJ. 2008;336(7646):701-704. doi:10.1136/bmj.39497.500903.25 [PubMed 18321957]
  69. Solomons RS, van Toorn R, Cresswell FV, Seddon JA. Update on the treatment of pediatric tuberculous meningitis. Pediatr Infect Dis J. 2022;41(9):e393-e395. doi:10.1097/INF.0000000000003557 [PubMed 35421048]
  70. 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.
  71. Streptomycin [prescribing information]. Big Flats, NY: X-Gen Pharmaceuticals Inc; October 2022.
  72. Traynor AM, Nafziger AN, Bertino JS Jr. Aminoglycoside dosing weight correction factors for patients of various body sizes. Antimicrob Agents Chemother. 1995;39(2):545-548. doi:10.1128/AAC.39.2.545 [PubMed 7726530]
  73. US Department of Health and Human Services (HHS) Panel on Opportunistic Infections in HIV-Infected Adults and Adolescents. Guidelines for the prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. Updated November 2019. Accessed January 15, 2020.
  74. US Department of Health and Human Services (HHS). 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/hiv-clinical-guidelines-adult-and-adolescent-opportunistic-infections/whats-new. Updated January 18, 2023. Accessed May 26, 2023.
  75. US Department of Health and Human Services (HHS). Guidelines for the prevention and treatment of opportunistic infections in children with and exposed to HIV. https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-pediatric-opportunistic-infections/whats-new. Updated September 2, 2022. Accessed May 26, 2023.
  76. Van Scoy RE, Wilkowske CJ. Antituberculous agents. Mayo Clin Proc. 1992;67(2):179-187. doi:10.1016/s0025-6196(12)61320-2 [PubMed 1347579]
  77. Verpooten GA, Giuliano RA, Verbist L, Eestermans G, De Broe ME. Once-daily dosing decreases renal accumulation of gentamicin and netilmicin. Clin Pharmacol Ther. 1989;45(1):22-27. doi:10.1038/clpt.1989.4 [PubMed 2910634]
  78. World Health Organization (WHO). Guidance for Health Workers: Treatment of Mycobacterium ulcerans Disease (Buruli Ulcer), 2012. http://apps.who.int/iris/bitstream/10665/77771/1/9789241503402_eng.pdf. Accessed July 8, 2015.
  79. World Health Organization (WHO). WHO Guidelines on Tularemia. 2007. http://whqlibdoc.who.int/publications/2007/9789241547376_eng.pdf. Accessed June 29, 2012.
  80. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the eleventh WHO model list of essential drugs. http://www.who.int/maternal_child_adolescent/documents/55732/en/. Published 2002.
  81. 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.
  82. World Health Organization (WHO). WHO consolidated guidelines on tuberculosis: Module 5: management of tuberculosis in children and adolescents. World Health Organization; 2022. https://www.who.int/publications/i/item/9789240046764. Published September 21, 2022. Accessed May 26, 2023.
  83. Yu PA, Tran EL, Parker CM, et al. Safety of antimicrobials during pregnancy: a systematic review of antimicrobials considered for treatment and postexposure prophylaxis of plague. Clin Infect Dis. 2020;70(suppl 1):S37-S50. doi: 10.1093/cid/ciz1231 [PubMed 32435799]
  84. Zhu M, Burman WJ, Jaresko GS, Berning SE, Jelliffe RW, Peloquin CA. Population pharmacokinetics of intravenous and intramuscular streptomycin in patients with tuberculosis. Pharmacotherapy. 2001;21(9):1037‐1045. doi:10.1592/phco.21.13.1037.34625 [PubMed 11560193]
Topic 10238 Version 214.0

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