You are on page 1of 20

Aminoglycosides

25  James E. Leggett

SHORT VIEW SUMMARY


• Usual intravenous (IV) adult dose: 7 mg/kg unmeasurable; typically, two serum furosemide, IV radiocontrast agents, and
every 24 hours for gentamicin, netilmicin, and concentrations in postdistribution phase (≥1 vancomycin
tobramycin or 20 mg/kg every 24 hours for hour after administration, separated by ≥1.5 • Indications: atypical mycobacterial infections,
amikacin half-lives) brucellosis, cholangitis, cystic fibrosis,
• Usual dose for synergy against gram-positive • Cerebrospinal fluid penetration: low diverticulitis, endocarditis, endophthalmitis,
infections: 3 mg/kg/day gentamicin every 8, • Common adverse effects: nephrotoxicity, meningitis, pelvic inflammatory disease,
12, or 24 hours ototoxicity plague, health care–associated pneumonia,
• Renal failure: 60% dose reduction • Contraindications: hypersensitivity synergy for gram-positive infections, tularemia,
• Therapeutic drug monitoring: not always • Drug-drug interactions: increased urinary tract infections
necessary for brief administration (<5 to 6 nephrotoxicity risk with concomitant
days); trough concentrations should be amphotericin B, clindamycin, foscarnet,

Aminoglycoside antibiotics have been an important part of the anti- CHEMISTRY


bacterial drug arsenal since the 1940s, when systematic screening of All aminoglycosides have an essential six-membered ring with amino
soil actinomycetes for the elaboration of antimicrobial substances group substituents, hence the name aminocyclitol.1 The descriptor ami-
yielded streptomycin. Produced by a species of Streptomyces, it was the noglycoside results from the glycosidic bonds between the aminocycli-
first antibiotic in the aminoglycoside family (Table 25-1)1 to be derived, tol and two or more amino-containing or non–amino-containing
directly or indirectly, from Streptomyces spp. (those aminoglycosides sugars. Spectinomycin differs in that it has an aminocyclitol ring but
with names ending in -mycin) or from Micromonospora spp. (those no amino sugars and no glycosidic bonds. The central aminocyclitol
with names ending in -micin). Neomycin, kanamycin, and gentamicin for streptomycin is streptidine, whereas for all other current aminogly-
are fermentation products with two or three chemical constituents. cosides, it is 2-deoxystreptamine. The standard numbering convention
Amikacin, netilmicin, dibekacin, and isepamicin are semisynthetic is illustrated in Figure 25-1. The aminocyclitol ring is numbered coun-
derivatives of the natural product. All aminoglycosides share similar terclockwise, and the linked sugar molecules clockwise. Figure 25-1
physical, chemical, and pharmacologic properties. also illustrates the structural basis of the aminoglycoside subgroups.
They demonstrate concentration-dependent killing and prolonged Neomycin and paromomycin are derived from Streptomyces spp. and
postantibiotic effects against susceptible organisms.2,3 Several members link to cyclic sugars at positions 4 and 5 of 2-deoxystreptamine. Both
of the aminoglycoside family have predictable in vitro activity against drugs contain a distinctive pentose linkage as well as linkage to two
Pseudomonas aeruginosa and most other aerobic gram-negative bacilli. hexose sugars. Among the commonly used aminoglycosides, neomy-
Some aminoglycosides have useful activity against mycobacteria; one, cin contains the largest number of free amino groups (six). The neo-
paromomycin, has been used to treat selected colonic protozoan mycin family comprises framycetin, neomycin, and paromomycin. All
pathogens, and a related antibiotic, spectinomycin, has been used to are too toxic for parenteral use. Framycetin is available outside of the
treat infections with Neisseria gonorrhoeae. The aminoglycoside anti- United States.
microbial activity may be additive to or synergistic with that of Kanamycin, tobramycin, amikacin, arbekacin, and dibekacin com-
β-lactams against infection by aerobic gram-negative bacilli or aerobic prise the kanamycin family (Table 25-2). All derive from Streptomyces
gram-positive cocci. Although most studies have failed to demonstrate spp. and link to cyclic sugars at positions 4 and 6 of 2-deoxystreptamine.
improved outcomes in patients treated with antibiotic combinations Tobramycin is 3′-deoxykanamycin B. Amikacin is kanamycin A with
compared with monotherapy,4,5 those in septic shock receiving combi- the semisynthetic addition of 2-hydroxy-4-aminobutyric acid to the
nations including aminoglycosides have shown reduced mortality in a amino group at position 1 of the aminocyclitol. Arbekacin and dibeka-
recent trial.6 The prevalence of aminoglycoside resistance has remained cin are available for clinical use outside of the United States.
low,7 and emergence of bacterial resistance during therapy has been Gentamicin is a mixture of C1, C1a, and the enantiomers C2 and C2a
rare. Mean aminoglycoside use in U.S. academic centers has declined elaborated by Micromonospora spp. with glycosidic linkages at posi-
by 41% from 2002 to 2009, with concomitant significant and opposite tions 4 and 6. Sisomicin is the dihydro analogue of gentamicin C1a, and
change in rates of resistance for some organisms and no change for netilmicin is derived from sisomicin by the addition of an ethyl group
others.7 to the amino group at position 1 of the aminocyclitol. Isepamicin is
The members of the aminoglycoside family share the potential for not approved for use in the United States. Aminoglycosides are highly
nephrotoxicity, ototoxicity, and, rarely, neuromuscular blockade. The soluble in water and insoluble in organic solvents.10 The latter property
risk of toxicity may be decreasing as mechanisms are understood, new correlates with the limited ability of aminoglycosides to cross lipid-
dosage strategies introduced, concomitant risk factors avoided, and containing cellular membranes. Aminoglycosides have a molecular
shorter drug courses used. Allergic reactions are rare. Because of patent size in the range of 445 to 600 daltons.10 The molecular structure is
expirations, the cost of many aminoglycosides is low. Although new unchanged by freezing, by heating to 100° C for up to 4 hours, or by
β-lactams and fluoroquinolones share the same antibacterial spec- changes in solution pH ranging from 3.0 to 12 over several hours.11-13
trum, the efficacy of the aminoglycosides and resistance problems with The pKa values of the individual amino groups can be determined by
the newer drugs presage a continued need. Recent reviews have indeed nuclear magnetic resonance spectroscopy.14 The overall pKa for genta-
reported that aminoglycoside use is once again increasing because of micin is about pH 8.4.10 Hence, at pH 7.4, the aminoglycosides have a
emerging gram-negative resistance to other available drugs.8,9 very high positive charge and are cationic.
310
310.e1
KEYWORDS
amikacin; aminoglycosides; antimicrobial therapy; combination
therapy; gentamicin; isepamicin; kanamycin; neomycin;

Chapter 25  Aminoglycosides


nephrotoxicity; netilmicin; ototoxicity; paromomycin;
pharmacodynamics; protein-synthesis inhibitors; tobramycin
311

TABLE 25-1  The Family of Aminoglycosides in Clinical Use*


GENERIC NAME PROPRIETARY NAME SOURCE YEAR REPORTED CHEMISTRY

Chapter 25  Aminoglycosides


Streptomycin None Streptomyces griseus 1944 Unique central aminocyclitol ring
Neomycin Mycifradin, Neobiotic Streptomyces fradiae 1949 Roughly equal proportions of neomycin B and C
Kanamycin Kantrex Streptomyces kanamyceticus 1957 Mixture of 95% kanamycin A and 5%
kanamycin B
Paromomycin Humatin Streptomyces fradiae 1959 Part of “neomycin” family
Spectinomycin Trobicin Streptomyces spectabilis 1961 Chemically distinct but closely related to
aminoglycosides
Gentamicin Garamycin Micromonospora purpurea and 1963 Roughly equal proportions of gentamicin C1, C1a,
Micromonospora echinospora and the enantiomers C2 and C2a
Tobramycin Nebcin Streptomyces tenebrarius 1967 Natural 3′-deoxy derivative of kanamycin B
Sisomicin† Siseptin Micromonospora inyoensis 1970 Dehydro analogue of gentamicin C1a
Dibekacin*,† Streptomyces kanamyceticus 1971 Dideoxy derivative of kanamycin B
Amikacin* Amikin Streptomyces kanamyceticus 1972 Semisynthetic derivative of kanamycin A
Netilmicin* Netromycin Micromonospora inyoensis 1975 N-Ethyl derivative of sisomicin
Isepamicin*,† Micromonospora purpurea 1978 I-N-S-α-Hydroxy B amino propionyl derivative of
gentamicin B
*Semisynthetic aminoglycosides.

Approved for human use in countries other than the United States.
From Wright GD, Berghuis AM, Mobashery S. Aminoglycoside antibiotics: structures, functions, and resistance. In: Rosen BP, Mobashery S, eds. Resolving the Antibiotic
Paradox: Progress in Understanding Drug Resistance and Development of New Antibiotics. New York: Plenum; 1998.

Streptidine NH2
2-Deoxystreptamine
CH NH
NH NH2
2 NH6 4 2
HO 1 3
H2N C N 3 A OH HO A NH2
4 OH HO 6
H 5 5 1
OH OH
6′ CH2NH2
NH2 4′
5′ O R1
HO B 6′ NHR2
CH NH HO 1′ 4 NH2 4′ C
3′ 2′ 2 5′ O AAC (3)
NH NH6 NH2 3
2 O A NH2 HO B
HO 1 5′ NH2
6 HO 1′ 4
H 2N C N 3 A OH R1 HOCH2 O 5 1 3′ 2′
3 2
OH OH 6′ CH2NH2 OH2 NH2
H 4
5 5′′ O OH O A
4′
O 4′
C
1′ 5′ O HO
4′ O R2 HO 4′′
5′′ 5 6 1
1′
4′′ 1′′ 3′ 2′ B NH2 O
5′ CHO OH HO 1′ 4 H3C
B 2′′ 3′ 2′ 2 C
H3C 3′ HO OH OH 3 1′′
2′
3′′ O O A NH2 H3CHN 3′′ 2′′
6′′ OH
HO O NH2 Neomycin 4′′ CH2OH HO 5 6 ANT(2′′)
O
5′′ O 1
6′′ 5′′ Gentamicin
HOH2C2 1′′ 6′′ HO C
O CH2NH2 H2N
C 3′′ 2′′
HO NCH3 4′ OH
3′′ 2′′ 5′ O O
4′′
OH H HO B NH2
HO 1′ 4 Kanamycin
Streptomycin 3′ 2′ 3 2
NH2
NH2 6′
CH3 5′ O A AAC (6′)
6 6′
CH2NH2 CH2NH2
OH H HOCH2 O 5 1 4′
H H OH 5′ O 5′
O
H2N+ O O CH3 5′′ O 4′ 1′ HO B 4′
B O NH2
CH2NH2 C HO 1′ 4 NH2 1′ 4 3 2
2′ 3′ 2′ 2 2′
6′′ OH
4′′ 1′′ 3′ 3 NH2
OH D O
3′
OH A NH2
_ CH2OH HO A NH2 HO
O HO O OH 4′′ O 6
HO 3′′ 2′′
4′′
5′′ O 5 6
H3C O 5 1
1
H OH Paromomycin C C
O NH2 HO 1′′ H3CHN
1′′
H3CNH2 H2N 3′′ 2′′
+ 3′′ 2′′ OH OH
O
2Cl- . 5H2O AAC (6′)
O Sisomicin
6′
CH2NH2 Tobramycin
4′
Spectinomycin 5′ O 6′
hydrochloride HO B CH2NH2
HO 1′ 4 NH2 O
3′ 2′ 2 5′
6′′ OH
3
O 4′
B O NH2
CH2OH HO A NHCCHCH2CH2NH2 1′ 4 2
4′′ 3
6 3′
5′′ O 5 1 NH A
HO C OH 2 HO NHCH2CH3
1′′ OH 4′′ 5′′ O 6
H 2N 3′′ 2′′ Amikacin AAC (2′) H3C O 5 1
OH C
O 1′′
H3CHN
3′′ 2′′ OH
O
Netilmicin
FIGURE 25-1  Chemical structure of the aminoglycosides and spectinomycin. Neomycin contains approximately equal amounts of neomycin B
(R1 = H; R2 = CH2NH2) and neomycin C (R1 = CH2NH2; R2 = H). Kanamycin is principally kanamycin A, as shown. Gentamicin is gentamicin C complex
with roughly equal amounts of C1 (R1 = R2 = CH3), C1a (R1 = R2 = H), and C2 (R1 = CH3; R2 = H). The sites of action of four inactivating enzymes are shown,
including three acetyltransferases—Aac(3′), Aac(2′), and Aac(6′)—and one adenyltransferase, Ant(2″).
312

TABLE 25-2  Chemical Families, Names, and Worldwide Availability


EXAMPLE AVAILABLE IN AVAILABLE OUTSIDE
FAMILY MEMBERS TRADE NAME UNITED STATES UNITED STATES
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

Streptomycin Streptomycin Streptomycin Yes Yes


Kanamycin Amikacin Amikin Yes Yes
Arbekacin Habekacin No Yes
Dibekacin Dikacine No Yes
Kanamycin Kantrex Yes Yes
Tobramycin Nebcin Yes Yes
Gentamicin Gentamicin (C1, C1a, C2, C2a) Garamycin Yes Yes
Isepamicin Isepacine No Yes
Netilmicin Netromycin No Yes
Sisomicin Sisogen No Yes
Neomycin Framycetin Soframycin No Yes
Neomycin Mycifradin, Neobiotic Yes Yes
Paromomycin Humatin Yes Yes
Spectinomycin* Spectinomycin Trobicin No Yes
*An aminocyclitol, no glycosidic bonds.
U.S. Food and Drug Administration approved but not available in United States.

The overall positive charge contributes to both antimicrobial activ-


ity and toxicity. Antibacterial activity is enhanced in media with an
alkaline pH and reduced in media with an acidic pH.13 The positively
charged aminoglycosides bind to the negatively charged RNA back-
bone, cell wall lipopolysaccharide (LPS), cell membrane phospholip-
ids, and other anionic molecules.15 G1405 U1495
Cationic aminoglycosides interact chemically with β-lactam
antibiotics.16-18 The reaction results in a nucleophilic opening of the
β-lactam ring, with acylation of an amino group of the aminoglycoside
and mutual loss of antibacterial activity. In vitro, gentamicin and tobra-
mycin are inactivated with greater ease than netilmicin, amikacin, or
isepamicin. Perhaps because their dosage is in grams rather than mil- III II
ligrams, the antipseudomonal penicillins (carbenicillin, ticarcillin,
piperacillin, mezlocillin, and azlocillin) are the β-lactams most suscep-
tible to the reaction. The reaction requires several hours in vitro, so the
clinical import is limited. In general, penicillins and aminoglycosides
IV
should not be mixed in the same solution before infusion, although a III
recent product reformulation does allow piperacillin/tazobactam I
co-administration with gentamicin at specific doses.19 Serum speci-
mens for drug concentration determination from patients receiving
both drugs should be assayed immediately or frozen. If patients with
renal failure are given an aminoglycoside and an antipseudomonal
penicillin concomitantly, there is a 10% to 20% reduction in the serum A1492
aminoglycoside concentration compared with the levels observed
when each drug is administered alone.20

MECHANISMS OF ACTION
Both electrostatic interactions between positively-charged aminogly-
coside amino groups and negatively-charged RNA phosphate groups, FIGURE 25-2  Three-dimensional structures of the aminoglycoside
as well as hydrogen bonds between multiple amino and hydroxyl antibiotics paromomycin (yellow) and gentamicin C1a (red) bound
groups of both, contribute to production of a tightly bound complex to different sections of the Escherichia coli 16S ribosomal RNA
that derails translation.21 Deoxystreptamine rings I and II, conserved (shown in light tan and dark tan, respectively). Rings I and II of the
and sequence specific among most aminoglycosides, are essential for two drugs, which are required for antibiotic activity, bind in the same mode
binding to the ribosomal decoding A site. Aminoglycoside antibiotics in both complexes; the additional rings (III and IV in paromomycin and III
preferentially bind with high avidity to a region of highly conserved in gentamicin C1a), which are less important for drug activity, make diverse
nucleotides at the A (acceptance) site22-24 in the 16S reverse transfer contacts with the RNA. (Courtesy Joseph D. Puglisi, Director, Stanford
Magnetic Resonance Laboratory, Stanford University School of Medicine.)
RNA portion of the messenger RNA (mRNA) decoding region of the
30S subunit of prokaryotic ribosomes.21,25 Aminoglycoside binding
induces a conformational change in three adenine residues that reduces
the fidelity of normal mRNA translation and translocation (Fig. of binding varies with the aminoglycoside, depending upon the
25-2),25,26 leading to the accumulation of truncated or nonfunctional number of amino groups and their state of protonation.14
proteins in bacteria. The respective nucleotide sequence differences Permeability of the polycationic aminoglycosides is enhanced in
between bacterial and human ribosomal subunits leads to lower affin- aerobic bacteria that use membrane-bound electron transporters.
ity of aminoglycosides for the latter. In eukaryotic cytosolic ribosomes, Initial electrostatic binding of aminoglycosides to the cell surface is
adenosine is replaced by a guanosine within the A site, with a concomi- followed by two energy-dependent uptake phases and binding to
tant reduction in binding of aminoglycosides. The double-adenosine ribosomes.28,29,30 In gram-negative bacteria, the cationic aminoglyco-
nucleotides in prokaryotic ribosomes create an internal bulge and a sides rapidly bind to negatively charged residues in the LPS, polar
larger groove that allows access to the ribosomal binding site.27 Avidity heads of phospholipids, and anionic outer membrane proteins.31-34 By
313
2+
competitively displacing cell wall magnesium ion (Mg ) and calcium the membrane. Therefore, anaerobic bacteria are intrinsically resistant
ion (Ca2+) bridges that normally link adjacent LPS molecules,35,36 ami- to aminoglycosides.
noglycosides result in a rearrangement of LPS with subsequent bleed- Low-level aminoglycoside resistance attributed to impaired cell
ing of the outer membrane, formation of transient holes in the cell wall, wall permeability may be the result of drug efflux mechanisms. Multi-

Chapter 25  Aminoglycosides


and disruption of the cell wall’s normal permeability function.37 drug (including aminoglycoside) efflux pumps are adenosine
After initial binding, aminoglycosides are transported across the triphosphate–dependent active pumps recently recognized as major
bacterial cytoplasmic membrane by a slow energy-dependent phase, contributors to antibiotic resistance.21 Constitutive expression results
EDP-I, that transports the drug into the cytosol and then bind to the in intrinsic low-level resistance; overexpression of these transporters
ribosome in a subsequent rapid energy-dependent phase, EDP-II.* The may result from regulatory gene mutations or increased antibiotic sub-
onset of bacterial killing is coincident with the transition from EDP-I strate concentration. Examples include the resistance nodulation cell
to EDP-II.29,30 Most bacterial cells manifest lethal injury after only 25% division (RND)-type superfamily in P. aeruginosa and the major facili-
of the maximum EDP-II uptake.39 The higher the external concentra- tator superfamily (MFS) in Escherichia coli.21,60 Activation of the
tion of aminoglycoside, the quicker the intracellular drug concentra- MexXY efflux pump also appears to explain adaptive resistance, defined
tion reaches a level necessary to trigger EDP-II uptake, which in turn as transient resistance to aminoglycosides that follows the rapid, early,
forecasts the death of the organism.40 concentration-dependent killing of susceptible bacteria.32,57 The refrac-
Binding of aminoglycosides to prokaryotic ribosomes is a prereq- tory state lasts beyond the postantibiotic effective period into the time
uisite for the drugs’ antimicrobial activity. However, the exact mecha- of regrowth. Adaptive resistance has been documented in vitro in
nisms of bactericidal activity remain unknown.1,21 The binding to animal models and in patients with CF,58,65,66 where the predominant
ribosomes is reversible, which usually results in a bacteriostatic rather identified mechanism of aminoglycoside resistance is efflux by the
than a bactericidal effect.1 Ribosomal binding results in a measurable MexXY-OprM system.21,67 Exposure of susceptible bacteria to amino-
decrease in protein synthesis as a result of misreading of mRNA. glycosides can select a second type of drug resistance. A subpopulation
Chloramphenicol and other drugs that inhibit protein synthesis are results from small colony variants with deficient energy-dependent
bacteriostatic and not bactericidal, which suggests additional unidenti- uptake of aminoglycosides and may result in clinical treatment failure.68
fied mechanisms of bactericidal activity. Recent work suggests that The least common mechanism involves alteration of the 16S ribo-
only bactericidal drugs stimulate hydroxyl radical formation in bacte- somal target of aminoglycosides. The best known example is resistance
ria as a function of metabolism-related depletion of reduced nicotin- of Mycobacterium tuberculosis to streptomycin as a result of point
amide adenine dinucleotide, destabilization of iron-sulfur clusters, and mutations in ribosomal protein S12 and in the 16S rRNA. Resistance
stimulation of the Fenton reaction.41 However, others have subse- of Mycobacterium abscessus and Mycobacterium chelonae to amikacin
quently shown that killing by antibiotics appears to be unrelated to is the result of a 16S rRNA point mutation. Methylating enzymes that
reactive oxygen species.42 modify the 16S rRNA and decrease drug-binding affinity, mostly
The mechanisms of the intracellular accumulation of high concen- located on transposons within transferable plasmids, have been
trations of drug and of cell death are unclear. It is suggested that the described in a growing number of aminoglycoside-resistant clinical
high intracellular concentrations result from aminoglycoside closure isolates worldwide.69-74
of voltage-gated channels, with subsequent trapping of drug.43 There The most common cause of aminoglycoside resistance is deactiva-
are many consequences of the accumulation of large concentrations tion by specific enzymes derived from bacterial genes originally encod-
of aminoglycoside inside bacterial cells, including binding to ribo- ing enzymes involved in normal cellular metabolism of both
somes with mistranslation of mRNA yielding abnormal proteins, loss gram-positive and gram-negative bacterial pathogens.1,21 Examples of
of cell membrane integrity with efflux of intracellular ions, and inhibi- sites of enzyme activity are shown in Figure 25-1. Enzyme modification
tion of DNA replication, in addition to stimulating hydroxyl radical results in loss of antibacterial activity.
formation.1,41 Three covalent modifications of aminoglycosides are recognized.1,75
The aminoglycosides have a variety of other biologic activities that The amino groups can be modified by N-acetyltransferases (AAC). The
are the subjects of ongoing study. Some 1800 genetic diseases arise, in hydroxyl groups can be modified by either O-nucleotidyltransferases
part, from base pair insertions, deletions, or substitutions that produce (ANT) or O-phosphotransferases (APH). The enzymatically modified
premature stop codons, resulting in production of incorrectly trun- drugs bind poorly to ribosomes and result in high levels of resistance.
cated, nonfunctional proteins.44 Examples include the transmembrane In addition, the aminoglycoside may bind directly to a modifying
conductance regulator protein of cystic fibrosis (CF), Duchenne mus- enzyme in lieu of the ribosomal target.76
cular dystrophy, Hurler syndrome, and nephrogenic diabetes insipidus. Specific enzymes are categorized by a nomenclature published in
Aminoglycosides can suppress premature stop codons and restore 1993.1,77 Each enzyme is described by its class (AAC, ANT, or APH),
physiologically active amounts of functional protein.45-51,52,53 The mech- a number in parentheses signifying the location of modification of the
anism of the suppression of the stop codons is under study; uracil and drug, and a Roman numeral indicating a unique aminoglycoside resis-
cytosine nucleotides are major determinants of optimal gentamicin- tance profile. Distinct genes resulting in identical resistance pheno-
induced readthrough.54 types are indicated by a lowercase letter after the Roman numeral. For
example, Aac(6′)-Ia describes an acetylating enzyme that modifies
MECHANISMS OF RESISTANCE aminoglycosides at the 6′ position (see Fig. 25-1); the resistance profile
Bacterial resistance to aminoglycosides results from some combination is the same as that of Aac(6′)-Ib, but the enzyme protein is unique. A
of three mechanisms: (1) reduction of intracellular aminoglycoside summary of modifying enzymes and their profile, source, and pheno-
accumulation resulting from bacterial membrane alterations that type is available elsewhere.1
reduce uptake and/or active efflux systems, (2) decreased aminoglyco- Modifying enzyme genes are spread by plasmids or transposons, or
side binding by mutation or methylation of the 16S ribosomal RNA both. Some are chromosomal. The plasmid-transposon genes can
(rRNA) binding site, and (3) enzymatic deactivation of aminoglyco- result in rapid spread of drug-resistant phenotypes both within and
sides by N-acetylation, O-nucleotidation, or O-phosphorylation.55-60,61 between bacterial species. The resistance genes in gram-negative
Aminoglycosides also induce bacterial biofilm formation, with associ- pathogens are diverse. In gram-positive pathogens, resistance is limited
ated bacterial cell wall surface adhesiveness. Biofilm formation is a to Aph(3′)-IIIa, Ant(6′), and a unique bifunctional enzyme known as
concern in the treatment of chronic infections, especially those with Aac(6′)-Aph(2″). In gram-negative organisms, a complex pattern of
foreign bodies.62 In addition, genomic studies in Pseudomonas aerugi- aac(6′)-I genes, combined with aac(3) and ant(2″) and others, is
nosa have suggested other novel mechanisms of resistance.63,64 For observed. The presence of the enzyme results in high-level resistance
aminoglycosides to enter a bacterial cell after initial electrostatic of gram-positive cocci to all aminoglycosides except streptomycin.
adsorption to the membrane, there must be an active electron trans- Similar genes have been described in amikacin-resistant gram-negative
port chain sufficient to generate an electrical potential difference across bacterial clinical isolates.78,79
All enterococci have intrinsic resistance to aminoglycosides, with
*References 28, 29, 30, 34, 38, 39. minimal inhibitory concentrations (MICs) ranging from 4 to 256 
314
µg/mL.80 The resistance is attributed to the facultative anaerobic Nonetheless, aminoglycosides are used successfully in the treatment of
metabolism of enterococci, which reduces the transmembrane poten- other intracellular infections, such as brucellosis, chronic forms of
tial and thereby limits drug intake. Concomitant exposure of entero- bartonellosis, tuberculosis, tularemia, and yersiniosis.94-96 Aminoglyco-
cocci to a cell wall–active drug such as ampicillin or vancomycin sides in combination with other drugs have been used successfully to
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

facilitates access of aminoglycosides to their ribosomal target site and treat infections with staphylococci, streptococci, enterococci, Listeria,
classic synergistic bactericidal activity. and mycobacteria. Susceptibility of mycobacteria varies with the
Acquisition of genes that encode aminoglycoside-modifying species. Streptomycin is the most potent aminoglycoside in vitro
enzymes leads to high-level aminoglycoside resistance and loss of syn- against M. tuberculosis, whereas amikacin is the most active against
ergistic activity with penicillins or vancomycin. At least nine genes Mycobacterium avium-intracellulare.
have been described that mediate resistance to aminoglycoside syner- The aminoglycoside paromomycin is too toxic for parenteral
gism in enterococci.81 The most important is the bifunctional gene administration. Because the drug is not absorbed from the intestinal
aac(6″)-Ie–aph(2″)-Ia, which encodes the bifunctional enzyme tract, it can be used safely as an alternative therapy for infection caused
Aac(6′)-Ie–Aph(2″)-Ia. A combination of resistance genes can result in by Entamoeba histolytica.95 In addition, a recent phase III trial provided
failure of synergism with all aminoglycosides available in the United evidence of its efficacy for ulcerative Leishmania major.97 Spectinomy-
States. Arbekacin, a derivative of dibekacin, available only in Japan, has cin was an alternative agent used intramuscularly in the treatment of
shown somewhat promising results in the presence of a variety of N. gonorrhoeae95 but is no longer distributed in the United States.
modifying enzymes.81 Urine is known to partially inhibit the activity of aminoglycosides
The current threshold for detection of high-level resistance to gen- against urinary tract pathogens. Inhibition is believed to result from
tamicin in vitro is an MIC of 500 µg/mL or greater82; for resistance to the low pH and high osmolality caused by the high salt and glucose
streptomycin, it is an MIC of 2000 µg/mL or greater. There are species concentrations. In addition, present data support the hypothesis that
differences in susceptibility to aminoglycosides. The MIC of tobramy- betaines, normally found in urine, permit the expression of increased
cin against Enterococcus faecium ranges from 62 to 1000 µg/mL; there aminoglycoside resistance.98
is no synergism with a cell wall–active drug. The difference is caused Three characteristics describe aminoglycoside antibacterial activity:
by an enzyme that modifies tobramycin but not gentamicin.83 concentration-dependent killing, the presence of a postantibiotic effect
Surveillance of the in vitro susceptibility of Enterobacteriaceae and (PAE), and synergism with other drugs.99 Aminoglycosides are rapidly
nonfermentative gram-negative bacilli demonstrates increasing resis- bactericidal, and their rate of bacterial killing increases as the antibiotic
tance to aminoglycosides as well as other classes of antibacterial drugs. concentration is increased, regardless of the inoculum.3 Exposure of
Because the multiclass resistance genetic elements are on mobile units, both gram-positive and gram-negative organisms to less frequent,
such as transposons or plasmids, their continued spread is anticipated. higher doses with in vitro pharmacokinetic models resulted in more
One or more of the mechanisms of aminoglycoside resistance (e.g., and faster killing initially, followed by regrowth of resistant mutants
enzymatic, target methylation, efflux pumps) are present in multidrug- and very similar 24-hour results, compared with more frequent dosing
resistant gram-negative bacilli. As a result of rapidly changing patterns regimens.100,101 Further studies indicated that emergence of resistance
of resistance, only general statements are possible regarding the antici- could be prevented with peak-to-MIC ratios greater than 8.102
pated in vitro and in vivo spectrum of antibacterial activity of In thigh infections of neutropenic mice, the rate of killing increases
aminoglycosides.84-90 with increasing mg/kg doses over the first few hours, whereas regrowth
is suppressed for several hours despite aminoglycoside levels less than
ANTIMICROBIAL ACTIVITY the MIC.103 The duration of this postexposure growth suppression, or
Although marked regional and individual hospital differences exist for PAE, also increases with higher doses.104,105
in vitro susceptibility patterns, the majority of aerobic and facultative The PAE represents suppression of bacterial growth after short anti-
gram-negative bacilli, including Enterobacteriaceae, P. aeruginosa, and microbial exposure.104,105 In vitro, the tobramycin-induced PAE against
Acinetobacter spp., in the United States remain susceptible to gentami- E. coli correlated with inhibition of protein synthesis but not of DNA
cin, tobramycin, and amikacin.7 Yersinia pestis is inhibited by strepto- or RNA synthesis.106 An aminoglycoside PAE can be demonstrated
mycin, and Francisella tularensis is inhibited by both streptomycin and after incubation with S. aureus but not after contact with S. pneu-
gentamicin. The aminoglycosides show no inhibitory activity against moniae.104 Inoculum size, oxygenation, and pH of the medium affect
Stenotrophomonas maltophilia or Burkholderia (Pseudomonas) cepacia. the duration of in vitro PAE.104,105,107 For the aerobic or facultative
Among gram-positive aerobic bacteria, methicillin-sensitive Staphylo- gram-negative rods tested, combinations with β-lactams other than
coccus aureus (MSSA) is susceptible and methicillin-resistant S. aureus imipenem result in the same PAE as that of the aminoglycoside
(MRSA) isolates are resistant. All streptococci, including Streptococcus alone.104,108 Rifampin was associated with synergistic enhancement of
pneumoniae, are resistant. Because the aminoglycosides require aerobic the PAE induced in P. aeruginosa by tobramycin.105,109
metabolism to exert an antibacterial effect, it is not surprising that all The in vivo PAE of aminoglycosides has been studied in at least five
the anaerobic bacteria are resistant to aminoglycosides. animal models.103,104,110 In neutropenic mouse thighs infected with 15
There are some minor differences in relative degrees of in vitro clinical isolates of Enterobacteriaceae, the in vivo PAE after gentamicin
potency of aminoglycosides. For strains of S. aureus, gentamicin and therapy varied from 1.4 to 6.9 hours. In the same model infected with
netilmicin show the most potency. The MIC or minimal bactericidal P. aeruginosa, increasing the dose of tobramycin fivefold increased the
concentration (MBC) of gentamicin against Serratia spp. is consis- PAE from 2.2 to 7.3 hours.103 The in vivo PAE is prolonged further in
tently twofold lower than that of the other aminoglycosides, and the renally-impaired or non-neutropenic mice and larger animals.111
MIC or MBC of tobramycin against P. aeruginosa is also consistently Aminoglycoside combinations have been widely studied in vitro
twofold lower than that of the other aminoglycosides. This increased and for many years have been used in combination with other antibiot-
potency increases both the peak/MIC and 24-hour area under the ics to enhance bacterial killing and improve clinical efficacy. Synergy,
curve (AUC)/MIC ratios. Although tobramycin is more active in a more than additive effect, has been shown in multiple time-kill curve
animal models of pneumonia, to date, no clinical efficacy data have experiments between an aminoglycoside and a cell wall–active antimi-
been presented that parallel these in vitro differences. Even though crobial (e.g., penicillin, cephalosporin, monobactam, carbapenem,
amikacin MICs are generally higher against Enterobacteriaceae than glycopeptide).2,99,112 The mechanism of aminoglycoside synergistic
gentamicin, amikacin remains active against most (>80%) strains, as activity may not be the same for all target organisms. Enhanced ami-
well as many P. aeruginosa strains that have acquired both gentamicin noglycoside uptake in the presence of a cell wall–active drug has been
and tobramycin resistance.13,91-93 demonstrated with enterococci, viridans streptococci, S. aureus, and
Aminoglycosides have in vitro activity against Haemophilus spp. P. aeruginosa.99 No combination of aminoglycoside and cell wall–active
and Legionella spp. but are not used clinically for infections with these drug is indicated as effective for MRSA. In vitro synergism against
organisms. Legionellae are intracellular pathogens, and the intracel- P. aeruginosa and Enterobacteriaceae99,113 does not appear to translate
lular antimicrobial activity of the aminoglycosides is hampered by into all clinical situations.114 Of equal import, the bactericidal activity
their low concentration in the acidic lysosomal compartment.94 of aminoglycosides can be antagonized by bacteriostatic agents such
315
99
as chloramphenicol and tetracycline. The mechanism is unclear. Pos- Five meta-analyses have shown statistically significantly improved
sibilities include inhibition of the energy-dependent uptake of amino- clinical outcomes, and three have shown significantly lower nephro-
glycosides and interference with movement of the ribosome along toxicity when aminoglycosides were dosed once daily. Likewise, equiv-
mRNA. Recent provocative modeling work suggests new innovative alence or a trend toward lower ototoxicity was shown with once-daily

Chapter 25  Aminoglycosides


combinations may likewise prove synergistic.115 administration. Among trials identifying a time to onset of toxicity,
Aminoglycoside combinations have been evaluated in animal once-daily dosing delayed nephrotoxicity compared with 8-hour
models of endocarditis, meningitis, pneumonia, peritonitis-bacteremia, dosing, until total duration approached 10 to 14 days when the inci-
pyelonephritis, osteomyelitis, myositis (mouse thigh), subcutaneous dence became similar in both groups. Two trials comparing once-daily
infection with or without a foreign body, and in additional condi- to twice-daily dosing of amikacin, gentamicin, and tobramycin showed
tions.99,105,116-121,122-125 In general, if the organism is susceptible to both that the incidence of nephrotoxicity was lower with once-daily dosing
the aminoglycoside and the companion drug, antibacterial activity is when using short courses of therapy and was delayed more for an
enhanced; virtually all studies showing effectiveness of combined equivalent total amount of standard therapy drug.189,190
therapy used a cell wall–active drug (β-lactam or glycopeptide) with Once-daily dosing regimens have similar or slightly better efficacy
the aminoglycoside. than twice- or three times–daily regimens. If current recommenda-
An aminoglycoside as part of combination therapy may prevent or tions limiting duration to only 5 to 6 days189-191 are followed, once-
delay the emergence of bacteria resistant to either the aminoglycoside daily dosing can also delay the onset of nephrotoxicity. For treatment
or the companion drug. In a series of studies, aminoglycosides were regimens lasting longer than 5 to 6 days, aminoglycoside dosing by
shown to reduce but not to fully prevent the emergence of quinolone- individualized pharmacokinetic modeling may result in less nephro-
resistant strains of Enterobacteriaceae or P. aeruginosa in a murine toxicity.192 Consideration should also be given to morning administra-
model of peritonitis.126-178 The concomitant use of an active β-lactam tion of aminoglycosides to minimize nephrotoxicity, when food intake
appears to prevent the emergence of gentamicin-resistant subpopula- and higher urinary pH in diurnally active humans may reduce renal
tions in neutropenic animals.129 toxicity.193
Treatment of infected animals with gentamicin markedly reduced The overview414 of the large, published, extensively analyzed
mortality but had virtually no influence on the incidence of abscess experience in patients indicates that once-daily aminoglycoside
formation.130-132 Treatment with clindamycin or metronidazole reduced administration
the incidence of abscess formation but had no effect on peritonitis- • Is as efficacious as the traditional multiple-dose method (see later
bacteremia or lethality.133 Combination therapy reduced both acute discussion of exceptions).
mortality and late abscess formation despite conditions adversely • May lower but not eliminate the risk of drug-induced nephrotoxicity
affecting aminoglycoside activity, such as low pH, at low oxygen and ototoxicity.
tension, and in the presence of drug-binding purulent debris.13,15,134 • Is simpler, less time consuming, and more cost-effective than
Data from infected animals and analysis of clinical trial data multiple-dose regimens.
support the correlation between high peak levels, increased dose, and • Probably should not be used in patients with enterococcal
antibacterial efficacy.121,135-137,138 Dose fractionation studies reduce the endocarditis.359
interdependence among the various pharmacokinetic parameters in • Needs further study in selected populations of patients, such as
small rodents with rapid aminoglycoside clearance. In these models, pregnant women, CF patients (see “Cystic Fibrosis”), patients with
the therapeutic efficacy of aminoglycosides correlates with the peak meningitis caused by aerobic gram-negative bacilli, and patients
serum concentration and the area under the concentration versus time with osteomyelitis.420
curve (i.e., AUC) over time.121,122 A compilation of clinical trials involv- • Does not worsen neuromuscular function even in critically ill, ven-
ing aminoglycosides for gram-negative bacillary infections has shown tilated patients; nonetheless, rapid intravenous infusion should be
that peak/MIC and 24-hour AUC/MIC ratios predicted outcome.137,139,140 avoided.421
Clinical efficacy increased from 55%, for a peak/MIC ratio less than or At present, once-daily aminoglycoside administration is as effica-
equal to 2, to 90%, for a ratio greater than 8, when four different ami- cious as traditional multiple-dose regimens and may reduce the risk of
noglycosides were all administered by identical traditional every-8- nephrotoxicity but needs further study in selected populations.
hour–dosing regimens. Efficacy improved from 47%, for tobramycin
AUC/MIC ratios less than 110, to 80%, for ratios greater than110. Both CLINICAL PHARMACOLOGY
peak greater than or equal to 10 and AUC greater than 150 correlated Aminoglycosides are generally administered intravenously (IV) over a
with rapidity of fever and leucotytosis resolution in gram-negative 30- to 60-minute period. However, outpatient slow push infusion over
pneumonia. These favorable results applied only to organisms with 3 to 5 minutes has been safely administered in more than 5000 patients
MICs less than or equal to 0.5 µg/mL. Current maximal dosing regi- over 15 years in one center.194 Aminoglycoside given intramuscularly
mens of 7 mg/kg routinely achieve peak/MIC ratios greater than or is absorbed completely, with maximal serum levels achieved between
equal to 10 for bacteria with MICs less than or equal to 1 µg/mL, but 30 and 120 minutes after administration.195 Absorption may be delayed
Clinical and Laboratory Standards Institute susceptibility breakpoints in the elderly and those patients with compromised renal function,
are 4 µg/mL. Synergistic activity with β-lactams might allow adequate hypotension, or impaired tissue perfusion. Aminoglycosides are mini-
coverage for those increasing numbers of organisms with MICs 2 to mally absorbed from the gastrointestinal tract.196 Nonetheless, instances
4 µg/mL.141,142 of deafness have resulted from administration of oral neomycin to
More than 55 clinical trials, including more than 30 prospective patients with hepatic encephalopathy and impaired renal function.197
randomized trials and 9 formal meta-analyses, have compared amino- Also, increased absorption in the presence of concomitant inflamma-
glycoside once-daily administration with traditional 8- or 12-hour tory bowel disease is of theoretical concern. In contrast, patients with
administration, showing equivalency or superiority of once-daily regi- acquired immunodeficiency syndrome and severe cryptosporidiosis
mens. In largely prospective fashion, more than 6500 mostly non- have ingested large amounts of paromomycin over protracted periods
neutropenic adult patients have received amikacin, gentamicin, without evidence of toxicity. Other exposures may lead to systemic
netilmicin, or tobramycin for periods of 7 to 14 days.143-185,186-188 toxicity. Topical application of aminoglycoside on inflamed skin leads
Comparable antimicrobial efficacy and toxicity were recorded for a to no or minimal absorption. However, patients with extensive burns
broad range of infections, including pneumonia, intra-abdominal or other severe dermal injury may absorb a drug and be at risk for
infections, gram-negative sepsis, febrile neutropenia, pelvic inflam­ toxicity.198 Aminoglycosides can be instilled into the pleural space or
matory disease, and urinary tract infections. Populations studied the peritoneal cavity; absorption is rapid, with resultant serum concen-
included the elderly,159,172 febrile neutropenic patients,† critically ill trations proportionate to the concentration of drug instilled. The use
patients,146,154,172,188 and patients with variable levels of renal of aminoglycosides in abdominal irrigation solutions is not recom-
insufficiency.152,172,183,186 mended because rapid absorption with subsequent neuromuscular
blockade has been reported.199 In contrast, aminoglycosides have

References 145, 160, 167, 172, 183, 187. been administered as a bladder irrigant, as an aerosol, and by direct
316
instillation into the lumbar sac or lateral ventricles without evidence TABLE 25-3  Typical Pharmacokinetic Parameters
of detectable concentrations in the blood.200-202 Inhalation has also been for Amikacin, Gentamicin, Netilmicin, and
used, especially in cystic fibrosis patients.203-207,208 Tobramycin in Adults with Normal Renal Function
As anticipated for drugs with a low level of protein binding (approx-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

imately 10%) and a high level of solubility in water, the aminoglyco- PARAMETER MEAN (± SED) OR RANGE
sides are distributed freely in the vascular space and relatively freely in Clearance (mL/min/kg)
the interstitial spaces of most tissues.209 Significant interpatient vari-   Creatinine ≤ 1.5 1.33 (0.61)
ability in aminoglycoside volume of distribution has been demon-   Creatinine > 1.5 0.53 (0.35)
strated, as well as intrapatient variation during therapy.210,211 In the   CrCl ≥ 100 1.51 (0.63)
absence of disease or infection, the volume of distribution mirrors that Volume of distribution (L/kg)
of the extracellular fluid compartment—0.2 to 0.3 L/kg.212 The volume   Dehydration 0.07-0.15
of distribution increases in edematous states, including ascites, in   Euvolemic 0.15-0.25
patients with burns, in patients with cystic fibrosis, and in some severe
  Expanded ECF 0.35-0.70
infections, when conditions may vary from hour to hour in critically
Half-life (hr)
ill patients.213 The volume of distribution increases less than expected
or actually decreases in obese individuals.214 Because of their size,   Creatinine < 1.5 0.15-15
polycationic charge, and lipid insolubility, aminoglycosides cross bio-   CrCl ≥ 100 0.5-7.6
logic membranes poorly, with the exception of renal tubular cells and   Age < 30 yr 0.5-3
perhaps inner ear cells that appear to have an inherent transport mech-   Age > 30 yr 1.5-15
anism. The cells of the renal proximal convoluted tubule can concen- Urinary excretion 85%-95%
trate aminoglycosides to levels that exceed those of plasma or interstitial CrCl, creatinine clearance; ECF, extracellular fluid; SED, standard error of the
fluid.215 difference.
Adequate antibiotic concentrations are achieved in most body Modified from Schentag JJ, Meagher AK, Jelliffe RW. Aminoglycosides. In:
fluids.216 Aminoglycosides enter synovial fluid easily, with subsequent Burton ME, Shaw LM, Schentag JJ, et al, eds. Applied Pharmacokinetics and
Pharmacodynamics: Principles of Therapeutic Drug Monitoring. Philadelphia:
concentrations only slightly less than simultaneous serum concentra- Lippincott Williams & Wilkins; 2006.
tions.217 Parenteral aminoglycoside administration results in low con-
centrations of active drug in bronchial secretions.202 Penetration into
epithelial lining fluid ranges from 32% to 54% of serum concentra-
tions.218,219 Much higher concentrations can be achieved by aerosol TABLE 25-4  Suggested Dosing Regimens for
administration.204 Diffusion is slower in bile, feces, prostatic, and amni- Adults
otic fluid.220-222 ESTIMATED DOSE (mg/kg)
Aminoglycosides traverse the blood–cerebrospinal fluid and blood- CREATININE Gentamicin, DOSING
brain barriers poorly223 but do cross the placenta and achieve fetal CLEARANCE Netilmicin, INTERVAL
serum concentrations 21% to 37% of maternal concentrations.222 Intra- (mL/min) Tobramycin Amikacin (hr)
ventricular administration results in high concentrations in both ven- 100 7 20 24
tricular and spinal fluid.223-226 Therefore, the intraventricular route is 90 7 20 24
recommended for meningitis caused by aerobic gram-negative bacilli 80 7 20 24
in adults in the rare cases in which this therapy is necessary. However, 70 5 15 24
in newborns, intraventricular aminoglycoside is no more effective and 60 5 15 24
perhaps more toxic than the drug given IV. Aminoglycoside penetra-
50 4 12 24
tion into the tissues of the eye is poor; neither systemic nor subcon-
40 4 12 24
junctival administration in single doses produces reliable levels in the
vitreous humor of humans.227-230 Direct intravitreal injection is recom- 30 5 15 48
mended for the treatment of endophthalmitis. 20 4 12 48
Urine concentrations of aminoglycosides exceed peak plasma levels 10 3 10 48
25- to 100-fold within 1 hour after drug administration.231,232 Because <10 2.5 7.5 48
of renal tubular cell absorption and subsequent release, urine concen- Modified from Gilbert DN, Bennett, WM. Use of antimicrobial agents in renal
trations remain above therapeutic levels for several days after a single failure. Infect Dis Clin North Am. 1989;3:517-531.
dose, even in severe renal impairment. After termination of a multiple-
dose regimen, urine levels remain above therapeutic levels for days,
with a terminal half-life of 48 to 200 hours.232-234 and AUCs are larger. Suggested initial dosing regimens for typical
Aminoglycosides are primarily eliminated unchanged by the kidney aminoglycosides in clinical use are listed in Table 25-4. One caveat to
via glomerular filtration. Less than 1% is eliminated in the feces and proper interpretation of these published nomogram concentrations is
1% in saliva.235 Aminoglycosides can be removed by dialysis, leading that the “peaks” were likely obtained before the end of distribution,
to unpredictable clearance depending upon the specific modality and whereas pharmacodynamic data uses real postdistribution peaks.
dialysis membrane capacity.236 With normal renal function in adults, Blind use of a 7-mg/kg dose may not achieve peaks related to expected
more than 90% of an administered dose is recovered in urine unchanged pharmacodynamic outcomes.241 Disease states that result in significant
during the first 24 hours.235,237 The remainder is slowly recycled to the loss of muscle mass are associated with low serum creatinine values.
tubular lumen, with a tissue half-life of 30 to 700 hours,237,238 creating In such patients, the Cockcroft-Gault formula may seriously overesti-
a multicompartmental elimination profile.239,240 mate the glomerular clearance of aminoglycosides.242 For this reason,
The pharmacokinetics of aminoglycosides in clinical use are very it has been suggested that the minimum value for subcutaneous (SC)
similar, even though interpatient variability is broad and associated administration used in the formula be 0.8 µg/mL.243 However, a recent
with body weight, body composition, and renal function as noted study in geriatric patients showed that such a practice significantly
above. Typical expected aminoglycoside pharmacokinetic parameter underestimated creatinine clearance (CrCl) and recommends that the
values are summarized in Table 25-3. Peak serum concentrations of practice should be avoided.244 The U.S. Food and Drug Administration
gentamicin, netilmicin, and tobramycin after a 7-mg/kg dose infused (FDA), several decades before pharmacodynamic research into dose
over 30 minutes range from 15 to 20 µg/mL, and yield an AUC of optimization, approved gentamicin and tobramycin for up to 1.7 mg/
70 to 100 mg/hr/L. Amikacin 15 mg/kg similarly infused produces kg per dose and three doses per day, for a total daily dose of 5.1 mg/
peak levels of 41 to 49 µg/mL and an AUC of 110 to 145 mg.hr/L.2 kg.245 For netilmicin, the total daily dose is 6.0 mg/kg, and for amika-
In patients with renal impairment, peak concentrations are higher cin, it is 15 mg/kg. In one clinical study that included a high percentage
317
of critically ill patients, the peak serum gentamicin concentration after Also important are untoward effects that are encountered rarely.
the first dose of 5.1 mg/kg was less than 16 µg/mL in 48% of the Hypersensitivity reactions are uncommon, and the aminoglycosides do
patients.245 Therefore, in critically ill patients, it is reasonable to give an not provoke inflammation. Hence, phlebitis at intravenous infusion
initial dose of 7.0 mg/kg.188 Extended-interval dosing is the norm sites is rare; intramuscular injection sites do not become painful; instil-

Chapter 25  Aminoglycosides


today, used in 75% of all acute care hospitals.246 No controlled studies lation into the pleural space, abdominal cavity, or cerebrospinal fluid
have compared 5 versus 7 mg/kg/day of gentamicin or tobramycin, but causes no irritation; and incorporation of an aminoglycoside into
clinical experience with the 7-mg/kg/day dosage is substantial.182,183 methyl methacrylate prosthetic joint cement is well tolerated over pro-
With the exception of patients with CF, the clinical use of single doses tracted periods. The aminoglycosides are not hepatotoxic, do not
of amikacin or netilmicin exceeding the licensed total daily dose has induce photosensitivity, and have no identified adverse influence on
not been reported. To date, FDA-approved labeling on therapeutic hematopoiesis or the coagulation cascade.
drug monitoring kits for aminoglycosides suggests “normal ranges” of
serum concentrations achievable with traditional three times–daily Nephrotoxicity
dosing rather than the higher, more effective serum concentrations Experimental Nephrotoxicity
now achievable with extended-interval dosing. The precise mechanisms of aminoglycoside-induced injury to renal
Various dialysis modalities clear differing amounts of aminoglyco- proximal tubular cells remain incompletely understood. There are as
sides, roughly one half of circulating aminoglycoside per hemodialysis yet unidentified genetic factors and major differences in susceptibility
period, depending on the characteristics of the dialysis membrane, to nephrotoxicity between animal species and between inbred strains
duration of dialysis, the patient’s blood pressure during dialysis, and of a specific animal.259
other variables. Depending on variables related to both the patient Aminoglycosides initially bind to an endocytotic receptor,
and the filter, continuous hemofiltration results in the equivalent of a megalin,260,261 important in the endocytotic uptake of proteins in the
CrCl of 10 to 50 mL/min.247,248 For patients undergoing continuous renal proximal tubule.260-262 In megalin receptor knockout mice, there
ambulatory peritoneal dialysis who have a systemic infection and are was no renal uptake of aminoglycosides.263 After binding to the anionic
receiving an intravenous dose of aminoglycoside every 2 to 3 days, it megalin in clathrin-coated pits,260,264 the cationic aminoglycoside is
is necessary to give small daily intravenous supplements to replace the then transferred to endosomes, and the megalin is returned to the
drug lost in the dialysate or dosed based upon seum concentrations apical plasma membrane. A portion of drug-containing endosomes
(www.pdiconnect.com/content/30/4/393.full.pdf+html). For patients fuse with lysosomes, where the aminoglycosides inhibit lysosomal
on hemodialysis, a traditional dose is given every 48 to 72 hours and, phospholipases, resulting in lysosomal whorl-like membrane changes
on the day of hemodialysis, an additional one half of the full dose is that, because of their morphologic appearance, have been termed
given, after dialysis, to replace drug that was removed by the dialysis myeloid bodies.260 Some of the aminoglycoside-containing endosomes
or simply dosed after dialysis. Because of individual variability, serum move rapidly to the Golgi apparatus.265 The time frame correlates with
levels should be measured.249-251 the observed rapid decrease in cellular protein synthesis after amino-
Dosing for the morbidly obese is generally based on excess body glycoside exposure.266-269
weight multiplied by 0.4 plus ideal body weight. More recent work In cells that internalize aminoglycosides, cellular necrosis occurs
indicates that lean body weight permits simplified aminoglycoside gradually, during which time a variety of abnormalities are demon-
dosing across all weight strata, with clearance best predicted by the strable,270-281 leading to activation of the apoptotic pathway, with sub-
chronic kidney disease–epidemiology (CKD-EPI) equation.252-254 sequent necrosis of the cells of the proximal tubules.281 Initially,
However, the best clinical descriptor of renal function for pharmaco- animals manifest nonoliguric renal failure.282 After several days, there
kinetic modeling in geriatric patients is the Cockcroft-Gault–based is a fall in glomerular filtration. The cells of the proximal tubule can
model.255 Monitoring early peak concentration (and another to esti- regenerate with return of glomerular filtration. Regeneration occurs
mate AUC) may assist in optimizing aminoglycoside efficacy.8,141 It even if there is continued administration of the aminoglycoside.283 In
remains unclear whether every patient administered an aminoglyco- animals, the regenerated tubular cells have a reduced capacity to take
side requires an individual pharmacokinetic evaluation,256 but indi- up aminoglycosides.
vidualized dosing is essential in the critically ill patient with altered A hierarchy of nephrotoxic potential exists among the
volume of distribution of drug and unstable renal function. Use of new aminoglycosides.284-286 Neomycin is the most toxic aminoglycoside,287
Bayesian analysis in routine clinical care holds promise.257 Overall, and streptomycin is the least nephrotoxic, perhaps because the drug
individualized dosing results in greater efficacy and reduced toxicity does not accumulate in the renal cortex.
when compared with the nomogram approach.244,255,258 Aminoglycoside uptake into the proximal renal tubular cell is satu-
Typically, two concentrations in the postdistribution phase (>1 rable at clinically relevant concentrations, governed by Michaelis-
hour after administration, separated by greater than 1.5 half-lives) are Menten kinetics.260 Thus, for a given total daily dose of a specific
used; trough levels should be unmeasurable. aminoglycoside, the magnitude of toxicity is greatest when the dose
With the exception of the aminocyclitol spectinomycin, aminogly- is divided into multiple small increments and least when it is given
coside antibiotics share the potential for causing injury to the renal as a single daily dose.285,288,289 In addition, age, sex, volume status,
proximal convoluted tubules, damage to the cochlea or vestibular pH, and electrolyte balance have all been examined, with older age,
apparatus or both, and neuromuscular blockade (Table 25-5). The dehydration, lower urinary pH, and electrolyte depletion associated
inherent toxicity and relative toxic potential of the aminoglycosides with increased toxicity.61-74,75-275,290-301 The impact of experimental liver
correlate with their positive electrical charge at physiologic pH.10 disease or diabetes is unclear, but accumulated clinical data indicate
that concomitant liver disease is a risk factor for nephrotoxicity.302-306
Various drugs influence the severity of experimental aminoglycoside
nephrotoxicity.307-315 Vancomycin and the related drug teicoplanin
TABLE 25-5  Estimated Frequency of Serious amplify experimental aminoglycoside nephrotoxicity.307-315 Extended-
Clinical Adverse Reactions after Administration spectrum penicillins lower the risk of kidney injury.310,311 Polymers of
of Aminoglycoside Antibiotics aspartic acid dramatically reduce renal tubular injury despite the accu-
ADVERSE ESTIMATED USUAL mulation of very high renal concentrations of aminoglycoside.316-325
REACTION FREQUENCY (%) REFERENCES Animal experiments with an aminoglycoside plus cephalothin, cefazo-
Nephrotoxicity 5-15 237-247 lin, or cefamandole indicated either no effect or an attenuation of
Ototoxicity 2-14 nephrotoxicity, compared with an aminoglycoside plus placebo.326 Fos-
  Cochlear 2-10 268-270
fomycin may reduce aminoglycoside experimental nephrotoxicity by
inhibiting gentamicin-induced lipid peroxidation.327 In general, ascrib-
  Vestibular 3-14 268, 296, 297
ing causation to a single factor in a multivariate interaction resulting
Neuromuscular blockade Exceedingly rare 135, 309
in toxicity is not a statistically valid approach. In a clinical study, no
318

TABLE 25-6  Factors That Increase Risk for preexisting decrease in the GFR, and was not a risk factor in several
Aminoglycoside Nephrotoxicity studies.329,330,334 Hypotensive patients, especially those with septic shock
or sepsis syndrome, have an increased incidence of renal insufficiency.
Patient-related factors The role of aminoglycosides is unclear in that infection-induced low
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

  Older patient
  Preexisting renal disease perfusion pressures, consumptive coagulopathy, cytokine-mediated
  Female200 endothelial damage, and other factors may be etiologic in the fall in
  Male245 the GFR.341,342 Liver disease was identified as a risk factor in the retro-
Volume depletion, hypotension spective analysis of two large clinical trials and was then validated in
Hepatic dysfunction
Aminoglycoside factors two additional prospective trials.304,339
  Recent aminoglycoside therapy Clinical trial data support the concept that several days of therapy
  Larger doses are needed to cause nephrotoxicity of clinical consequence. In an
  Treatment for ≥3 days observational study of patients treated with combination therapy,
  Drug choice (e.g., gentamicin)240
  Frequent dosing interval118,253 including an aminoglycoside for infective endocarditis, there was a
Concomitant drugs 0.5% decrease per day in the estimated endogenous CrCl.340 In con-
  Vancomycin261-263 trast, accidental massive overdosage of 1 day or less has not resulted in
  Amphotericin B acute tubular necrosis.343,344
  Furosemide
  Clindamycin The influence of concomitant drugs is difficult to interpret in
  Piperacillin patients with serious or complex disease states who are receiving many
  Cephalosporins?248,258-260 drugs. Nonetheless, most studies suggest an increased risk of a fall in
  Methoxyflurane266
  Foscarnet
the GFR when other nephrotoxic drugs are administered concomi-
Intravenous radiocontrast agents tantly with aminoglycosides.§ Three prospective studies, one of which
was double blind, found the combination of cephalothin plus amino-
Data from references 201, 202, 238, 245, and references cited in the table.
glycoside more nephrotoxic than that of a penicillin derivative plus
an aminoglycoside.349-351 Subsequent multiple logistic regression risk
factor analysis identified a variety of cephalosporins as risk factors.
identifiable risk factor alone or in combination reliably predicted These results are consistent with ceftazidime enhancement of gentami-
nephrotoxicity.328 cin enzymuria in healthy volunteers.352 Two studies evaluated con-
comitant vancomycin administration; one analysis included a control
Clinical Nephrotoxicity group that received only an aminoglycoside.353,354 Both studies indi-
Aminoglycosides accumulate in the kidney, accounting for 40% of the cated that vancomycin was a risk factor. In febrile neutropenic patients
total drug in the body,240 and nearly 85% of this is located in the renal administered gentamicin or tobramycin plus carbenicillin or ticarcil-
cortex. lin, the reported incidence of nephrotoxicity was 2% to 6%, compared
The reported incidence of nephrotoxicity varies from 0% to 50%, with 10% to 15% or higher when the aminoglycoside was combined
with most reports in the 5% to 15% range (see Table 25-5).329-336 The with other β-lactam antibiotics.349,350,355 Of interest, a risk factor analy-
variability results from differences in the definition of nephrotoxicity, sis found an increased risk with concomitant piperacillin but not with
the frequency of testing, and the particular tests used to measure renal carbenicillin or ticarcillin. The authors speculated that the lower
function and the clinical setting in which the drugs are administered. sodium content of piperacillin might explain the difference.311
Minimal clinical differences were seen in a survey of clinical trials
involving approximately 10,000 patients between 1975 and 1982.329 Ototoxicity
Frequencies of nephrotoxicity averaged 14.0% for gentamicin, 12.9% Aminoglycoside antibiotics can cause cochlear and vestibular damage
for tobramycin, 9.4% for amikacin, and 8.7% for netilmicin. In pro- in experimental animals and humans.356 Streptomycin-induced hearing
spective, randomized studies with definitions of nephrotoxicity that loss and dizziness were included in the first clinical report of the drug’s
reflect a substantive decrement in the glomerular flow rate (GFR) in efficacy.357 Overt ototoxicity was noted in 2% to 10% of patients in early
seriously ill patients, the reported incidence varied between 5% and clinical experience.358,359 Ototoxicity is of particular concern because it
10% of patients’ courses.331-334,335,336 is usually irreversible and can appear after the end of treatment; more-
In studies of the etiology of acute renal failure, medication-induced over, repeated exposure engenders cumulative risk.360 A given patient
renal injury is reported as a major cause. In an analysis of more than may suffer only cochlear damage or only vestibular damage, but in
2000 hospitalized patients, almost 100 experienced renal insufficiency, general, the predominant lesion is vestibular.361 Rarely, both organs are
and seven episodes were attributed to aminoglycoside therapy.337 In injured. It is unusual to have both ototoxicity and nephrotoxicity in the
general, the aminoglycoside-induced decrement in the GFR is small. same patient.
Most patients have a nonoliguric fall in CrCl; progression to dialysis-
dependent oliguric-anuric renal failure is rare. As in animal models, Cochlear Toxicity
the tubular injury is reversible, and in a few patients, recovery of renal Few recipients of aminoglycoside therapy complain of hearing loss,
function has been documented despite continued administration of and yet the reported incidence is as high as 62% when asymptomatic
the aminoglycoside.338 high-frequency audiograms are performed repeatedly.362 The overall
Reported clinical risk factors for aminoglycoside nephrotoxicity incidence has been reported to be between 3% and 14%.363
can be grouped as being related to the patient, to the aminoglycoside, Normal sound perception extends to frequencies of 20 kHz; per-
or to the influence of the selected concomitant drug (Table 25-6).‡ ception of human speech requires sound detection in the range of 0.3
Factors identified in clinical trials include older age, preexisting renal to 3 kHz. A loss of hearing threshold of 25 to 30 dB is necessary before
or liver disease, shock, larger volume of distribution, location in inten- the patient is aware of the deficit. A commonly used definition for
sive care, pneumonia, rapidly fatal prognosis, leukemia, longer dura- drug-induced ototoxicity is an increase in auditory threshold of 15 dB
tion of therapy, and concomitant vancomycin or other nephrotoxin. or greater at any of two or more frequencies.360
Female sex was identified as a risk factor in one study but not con- Controlled data on cochlear toxicity are sparse. In a series of pro-
firmed in others.292,329,334-336 Reported clinical studies with cephalospo- spective clinical studies that examined the efficacy and toxicity of gen-
rins did not include an aminoglycoside-only group of patients, so it is tamicin, tobramycin, and amikacin in combination with β-lactam
not possible to ascertain the effect of cephalosporins on the risk of antibiotics, 22% of the aminoglycoside recipients had documented
nephrotoxicity. The correlation of increased risk of toxicity with age audiometric toxicity compared with 7% of cefotaxime-treated
and with preexisting renal disease may be misleading. It is unclear patients.333 In a different study of 53 subjects administered gentamicin,
whether a risk exists when the dosing regimen is adjusted for a tobramycin, or amikacin for at least 4 days, loss was unilateral in 55%
‡ §
References 292, 293, 329, 330, 334, 339, 340. References 292, 293, 329, 330, 334, 345-348.
319
and bilateral in 45% of the patients, and hearing loss was initially blockade has been described in patients administered neomycin, strep-
detected after a mean of 9 days of therapy.363 tomycin, kanamycin, tobramycin, gentamicin, amikacin, or netilmi-
Aminoglycosides penetrate into endolymph and cochlear and ves- cin.199,414 In general, blockade has occurred in clinical situations in
tibular tissue364 cells either by endocytotic uptake or mechanoelectrical which a disease state or a concomitant drug interferes with neuromus-

Chapter 25  Aminoglycosides


transduction channels365 and lysosomal accumulation after trans-strial cular transmission.414-420 Drug exposure may have been a result of
trafficking.366-368 In experimental animals, aminoglycosides can be intraperitoneal, intravenous, intramuscular, intrapleural, oral, topical,
detected in inner ear fluid within 3 hours of administration but or retroperitoneal administration.199,414 Aminoglycoside therapy in
cochlear damage requires 3 weeks of daily injections.360,369,370 The ventilated patients or others in intensive care has not been associated
precise mechanism of cochlear toxicity has eluded detection.371-373 As with adverse effects.421,422
in renal tubular cells, there is evidence for aminoglycoside-induced Blockade results from inhibition of the presynaptic release of
apoptotic cell death in cells of both the organ of Corti and the vestibu- acetylcholine as well as blockage of postsynaptic receptor sites of
lar apparatus.374 Hair cell loss has been considered irreversible. acetylcholine.423-426
However, animal studies in nonmammals and mammals have docu-
mented potential regeneration.375-377 CLINICAL INDICATIONS
The greatest risk for cochlear toxicity may be genetic predisposi- The aminoglycosides are effective in the empirical treatment of infec-
tion.378 In numerous reports, deafness has developed in family members tions caused by or suspected to be caused by aerobic gram-negative
of multiple pedigrees after treatment with an aminoglycoside and has bacilli, including P. aeruginosa. Aminoglycosides have in vitro activity
been associated with five or more different mutations in the mitochon- against S. aureus, but resistant small colony variants may appear
drial 12S rRNA gene,379-386,387,388 which may result in greater binding within 24 hours unless a concomitant antistaphylococcal β-lactam
affinity. No genetic predisposition to aminoglycoside vestibular or or vancomycin is administered.427 Activity against Enterococcus spp.
renal toxicity has been identified. requires a concomitant active penicillin or vancomycin. Aminoglyco-
Human toxicity is related to the dose and duration of aminoglyco- sides have no practical activity against pneumococci or anaerobic
side therapy.360 Ototoxicity is related to the AUC of concentrations in organisms. For reasons of anticipated spectrum of activity or to achieve
cochlear endolymph, itself correlated with AUC in serum.389 Thus, the an additive or synergistic effect, aminoglycosides are often combined
same total daily dose will result in the same incidence of ototoxicity, with a β-lactam antibiotic, vancomycin, or a drug active against anaer-
independent of the frequency of dosing.176,177,179,356 Measurable differ- obic bacteria.
ences in the risk of cochlear toxicity between gentamicin, tobramycin, The efficacy of empirical aminoglycoside therapy has been docu-
amikacin, and netilmicin are minimal, and less than some chemothera- mented in published symposia describing the results of clinical trials
peutic agents.363,390-399,400 Prolonged therapy beyond 10 days, renal or that served as the basis for licensure and subsequent trials that com-
hepatic impairment, and prior exposure to aminoglycosides are major pared one aminoglycoside with another or with a β-lactam.‖ In febrile
risk factors.397-399,401,402 neutropenic patients, a high failure rate was experienced after mono-
Concomitant “loop” diuretics, vancomycin, and loud ambient noise therapy with an aminoglycoside; therefore, in such patients, the ami-
increase the risk of cochlear toxicity.360,394-396 The use of aspirin reduced noglycosides are administered in combination with a β-lactam
the incidence of ototoxicity from 13% to 3%.390,403 antibiotic active against aerobic gram-negative bacilli.355 Guidelines on
the treatment of the febrile neutropenic patient suggest avoidance of
Vestibular Toxicity aminoglycosides, if possible; instead, empirical monotherapy with a
The target of drug-related vestibular toxicity is the type I hair cell of carbapenem or broad-spectrum β-lactam is used.432
the summit of the ampullary cristae.404 The true incidence of vestibular Given that optimal peak/MIC and AUC/MIC pharmacodynamic
toxicity in ill patients is virtually impossible to determine. Because ratios are obtained only for organisms with MICs of 0.5 µg/mL or less
vestibular injury is bilateral and initially symmetrical, it can be com- with conventional doses of gentamicin, netilmicin, and tobramycin
pensated by visual and proprioceptive cues, so patients can suffer con- (2 µg for amikacin), and MICs of 1.0 µg/mL with 7-mg/kg doses,8 it is
siderable injury before the appearance of symptoms or clinical findings. not surprising that several reviews have demonstrated inferior clinical
Suspicion is raised at the bedside by complaints of nausea, vomiting, efficacy of aminoglycoside monotherapy in severe gram-negative
and imbalance.361,405 Visual blurring with head movement (i.e., oscil- infections compared with therapy with β-lactams and fluoroquino-
lopsia) may occur. Symptoms are exacerbated in the dark, when the lones.355,442 Although aminoglycosides have been used in combination
eyes are closed, with moving or uneven surfaces, and in other situa- with β-lactams and fluoroquinolones to enhance killing and improve
tions that block compensatory pathways. Nystagmus may be evident. clinical efficacy, most studies have failed to demonstrate better out-
Systematic surveillance of patients with electronystagmography is comes with combinations compared with monotherapy.443
seldom performed; in one clinical study using electronystagmographic
surveillance, abnormalities were demonstrated in 4% to 6% of patients Bacteremia
receiving gentamicin or amikacin.335,336 There are no data that compare, In contrast to most older studies,444 and a recent retrospective review
in a controlled fashion, the toxic potential of the commonly prescribed of P. aeruginosa bacteremia,114 other recent studies demonstrate
aminoglycosides. improved outcome in patients with septic shock and gram-negative
Vestibular injury can be unilateral or bilateral and mild or bacillary bacteremia treated with combination aminoglycoside plus
severe.361,405 Functional recovery, even with bilateral damage, was β-lactam.
reported to occur in up to 53% of patients at 10 days to 9 months after Combination therapy provided a greater degree of initial appro­
cessation of drug exposure.405-408 In addition to visual and propriocep- priate therapy than β-lactam monotherapy, broader coverage than
tive compensation, recovery may be due to hair cell regeneration, as fluoroquinolones, and improved outcomes even in neutropenic
demonstrated in animal models.390,409,410 The relevance of the latter to patients.445,446 Reduced endotoxin release seen with aminoglycoside
humans is unclear. therapy341,342 may contribute to the diminished early mortality observed
Vestibular hair cells are purposely damaged by gentamicin as with combination therapy in patients with septic shock. Combination
therapy for Meniere’s disease that fails to respond to conservative therapy in selected cases is endorsed in the most recent international
measures.411,412 Injection into the middle ear allows gentamicin to guidelines for management of severe sepsis.447
pass through the round window membrane, penetrate the labyrinth, A detailed analysis and a meta-analysis of use of aminoglycosides
and destroy hair cells. A single injection is reported to effect good in the treatment of bacterial endocarditis have been published.448,449
control of vertigo in 75% of patients, with minimal sensorineural The use of aminoglycosides in combination with a β-lactam or vanco-
hearing loss.413 mycin may be of benefit for patients with streptococcal or enterococcal
endocarditis. The standard dosage to achieve a synergistic effect is
Neuromuscular Blockade 1 mg/kg IV every 8 hours; newer regimens have used 12- and 24-hour
Neuromuscular blockade after aminoglycoside administration is a
rare but serious and potentially lethal adverse effect. Neuromuscular ‖
References 4, 5, 331, 358, 359, 428-441.
320
intervals.450 The risk of nephrotoxicity increased in parallel with the aminoglycoside therapy in adult patients (aged 18 to 37 years) with CF
duration of therapy.340 For viridans streptococcal endocarditis, three is 17%, which is roughly the same as the rate in non-CF patients.
times–daily, twice-daily, and once-daily regimens appear to be of equal However, the risk per course of treatment is 2% in CF patients, com-
efficacy in reported clinical trials.429,448,449-451 pared with 7.5% in patients without CF.471 It is unclear how CF might
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

The long-held practice of administering combination β-lactams protect against aminoglycoside cochlear toxicity.
plus aminoglycosides for staphylococcal bacteremia and endocarditis The Cochrane Database of Systematic Reviews provides updated,
has been challenged by recent investigators,452,453 who recommended periodic critical evaluation of the use of aminoglycosides and other
against the addition of low-dose gentamicin (1 mg/kg every 8 hours), anti-infectives in the treatment of pneumonia in patients with CF.
because of its added nephrotoxicity. Others have instead emphasized Examples include single versus combination intravenous antibiotic
the suboptimal dosing strategy used454 or emphasized the significant therapy, nebulized antipseudomonal antibiotics, once-daily amino­
earlier (2 days vs. 4 days) defervescence.455 Aminoglyocoside combina- glycoside therapy, and elective versus symptomatic antibiotic
tions remain the standard of care for enterococcal bacteremia and therapy.472-477
endocarditis, although dual β-lactam regimens are being used as In 10 placebo-controlled trials of nebulized antipseudomonal anti-
alternatives.456,457 biotics that included 758 patients,207 patients receiving treatment had
a 12% increase in forced expiratory volume in 1 second and a reduced
Pneumonia odds ratio (0.69; 95% CI, 0.5 to 0.96) of need for hospitalization. Over
Aminoglycoside use in pneumonia is typically reserved for hospital- time, the incidence of aminoglycoside-resistant P. aeruginosa was
acquired or hospital-associated gram-negative bacillary respiratory greater in the treatment group.
tract infections, including those seen in patients on ventilators or Within 2 hours after the dose, sputum concentrations fall to
with cystic fibrosis. Combination therapy with a β-lactam yields approximately 14% of the levels found at 10 minutes after inhalation.206
superior results458 compared with aminoglycosides alone, without Absorption into serum is low. The average serum drug concentration
generally improving outcome over β-lactam monotherapy for Entero- 1 hour after inhalation was 1.0 µg/mL, with a range of 0.2 to 3.0 µg/
bacteriaceae.459 The role of combination therapy for P. aeruginosa mL.208 Ototoxicity has not been reported, but transient tinnitus
pneumonia is still unclear, with one meta-analysis suggesting signifi- occurred in a few individuals during clinical trials. Nephrotoxicity has
cant mortality benefit459 but another recent large study showing no not been observed. Aerosol therapy presents advantages of higher local
additional benefit as long as the isolate was susceptible to more than and less systemic exposure, self-administration at home, and improve-
one agent.114 ment in lung function with a reduced burden of P. aeruginosa.
Aerosolized aminoglycosides, initially used to treat cystic fibrosis
exacerbations,203-207,208 have recently shown promise in chronic bron- PROPHYLAXIS
chiectatic infections460,461 and ventilator-associated pneumonias. Most Clinical practice guidelines for antimicrobial prophylaxis in surgery
of the infections studied have involved P. aeruginosa, and the inhaled have recently been updated.478 Gentamicin or tobramycin, 5 mg/kg IV,
aminoglycoside used in conjunction with a systemic β-lactam. Aero- is recommended as an alternative in patients with β-lactam allergy in
solized aminoglycosides were associated with improved clinical and genitourinary and gastrointestinal procedures. For patients with val-
microbiologic cure rates, with less nephrotoxicity.462-464 A large, ran- vular heart disease, prophylaxis is no longer recommended solely to
domized placebo-controlled trial is currently underway.463 prevent endocarditis. For patients with a known or possible enterococ-
cal urinary tract infection, it is reasonable to include drugs with anti-
Intra-abdominal Infections enterococcal activity in the perioperative regimen for gastrointestinal
Current guidelines on empirical therapy for intra-abdominal infec- or genitourinary procedures.479
tions of moderate or high severity do not recommend the combination The risk of infection after elective colorectal procedures was signifi-
of an aminoglycoside with metronidazole.431 This judgment is based cantly reduced by mechanical cleansing of the bowel plus oral admin-
on both the toxic potential of the aminoglycosides and the availability istration of, usually, neomycin and erythromycin or metronidazole in
of equally efficacious regimens. Two recent meta-analyses of more than addition to standard IV antibiotic prophylaxis in recent controlled
5000 patients (including more than 3000 enrolled in randomized con- trials.478,480 A recent controlled trial demonstrated that an oral selective
trolled trials) demonstrated clinical inferiority of aminoglycoside digestive decontamination containing gentamicin effectively eradi-
therapy (usually as clindamycin/gentamicin combination) to its com- cated carbapeneum-resistent Klebsiella pneumoniae gastrointestinal
parator, β-lactam, for intra-abdominal infections. Nephrotoxicity was carriage,481 and might be used in nosocomial outbreaks.482 On the other
seen more often with aminoglycoside therapy, but overall toxicity was hand, the safety and efficacy of topical gentamicin in cardiac surgery
equivalent, as were all-cause and attributable-to-infection mortality.465 have not been clearly established.478,483 Multiple tunneled-catheter
Alternative first-choice regimens include a β-lactam–β-lactamase antibiotic-lock studies, including a recent prospective randomized
inhibitor combination, (e.g., piperacillin-tazobactam) plus a fluoroqui- trial,484 have demonstrated reduced catheter-related infections, but
nolone or monotherapy with a carbapenem. emergence of resistant pathogens remains a concern.485

Urinary Tract Infections Spectinomycin and Gonorrhea


A recent systemic review and meta-analysis of randomized controlled Spectinomycin has not been available in the United States since 2006.
trials in nearly 2500 patients enrolled in 26 trials showed that amino- It was previously recommended for pregnant women, patients in areas
glycoside monotherapy was equally effective as comparators in terms of high fluoroquinolone-resistance prevalence, and in men having sex
of all-cause mortality and treatment failure.428 Few trials enrolled with men. It showed 98% efficacy in uncomplicated urogenital and
patients with sepsis. A higher rate of bacteriologic failure and nephro- anogenital infections but only 52% efficacy in gonococcal infections
toxicity was observed with aminoglycosides. The duration of therapy involving the pharynx, where it does not reach therapeutic concentra-
was not recorded in the report, so it is unclear if 5 to 7 days of therapy tions.435 It is not effective in the treatment of infections with Treponema
would provide equivalent clinical efficacy to longer courses, given the pallidum or Chlamydia trachomatis. The drug is neither nephrotoxic
presence of therapeutic aminoglycoside levels for most pathogens in nor ototoxic. It is an alternative therapy for patients who are allergic
urine for 72 hours or longer after a single dose. Intravesicular genta- to β-lactams and for those who are infected with resistant strains of
micin has been investigated, with anecdotal success, for recurrent uri- gonococci. No intravenous form of the drug is available. Alternatively,
nary tract infections in intermittently catheterized patients.466 gentamicin has been used to treat gonococcal urethritis in Africa, and
isolates in Europe appear to be similarly susceptible.486
Cystic Fibrosis
CF patients demonstrate altered aminoglycoside pharmacokinetics Aminoglycosides in Orthopedic Surgery
requiring much larger doses of drug to achieve therapeutic serum Antibiotic-impregnated cement is used with increasing frequency in
levels.467,468 The frequency of nephrotoxicity in CF patients is less than primary hip and knee arthroplasties, as well as revision procedures of
that in non-CF patients.469,470 The prevalence of hearing loss after infected total joint arthroplasties,478 for which the FDA has approved
321
premixed aminoglycoside in bone cement products. Incorporation appears favorable, but a recent multicenter evaluation failed to show
of larger amounts of antibiotic, usually gentamicin or tobramycin, reduction in the risk of infection.495 A recent review of 20 mostly
allows release of higher drug concentrations but may adversely affect uncontrolled reports of antibiotic-containing spacers did not allow
mechanical properties. Concentrations and properties vary among evaluation of whether such adjunctive therapy provided additional

Chapter 25  Aminoglycosides


producers.487-492 Persistence of bacterial growth as adherent biofilms benefits to systemic antibiotic therapy.496 Prospective trials in
remains a potential problem,427,491,493 and nephrotoxicity has been aminoglycoside-containing beads for established osteomyelitis and
reported.494 To date, prophylactic use in primary joint arthroplasty prosthetic joint-associated infections are needed.497

Key References 141. Drusano GL, Ambrose PG, Bhavnani SM, et al. Back to the
future: using aminoglycosides again and how to dose them
363. Govaerts PJ, Claes PH, DeHeyring PHV, et al.
Aminoglycoside-induced ototoxicity. Toxicol Lett. 1990;52:
optimally. Clin Infect Dis. 2007;45:753-760. 227-251.
The complete reference list is available online at Expert Consult.
142. Tamma PD, Cosgrove SE, Maragakis LL. Combination 371. Guthrie OW. Aminoglycoside induced toxicity. Toxicology.
therapy for treatment of infections with gram-negative 2008;249:91-96.
1. Wright GD, Berghuis AM, Mobashery S. Aminoglycoside
bacteria. Clin Microbiol Rev. 2012;25:450-470. 386. Veenstra DL, Harris J, Gibson RL, et al. Pharmacogenomic
antibiotics: structures, functions, and resistance. In: Rosen
173. Bailey TC, Little JR, Littenberg B, et al. A meta-analysis of testing to prevent aminoglycoside-induced hearing loss in
BP, Mobashery S, eds. Resolving the Antibiotic Paradox:
extended interval dosing versus multiple daily dosing of cystic fibrosis patients: potential impact on clinical, patient,
Progress in Understanding Drug Resistance and Develop-
aminoglycosides. Clin Infect Dis. 1997;24:786-795. and economic outcomes. Genet Med. 2007;9:695-704.
ment of New Antibiotics. New York: Plenum; 1998:27-69.
174. Ali MZ, Goetz B. Meta-analysis of the relative efficacy and 398. Moore RD, Smith CR, Lietman PS. Risk factors for the
2. Craig WA. Optimizing aminoglycoside use. Crit Care Clin.
toxicity of once-daily versus multiple daily dosing of ami- development of auditory toxicity in patients receiving ami-
2011;27:107-121.
noglycosides. Clin Infect Dis. 1997;24:796-809. noglycosides. J Infect Dis. 1984;149:23-30.
4. Paul M, Benuri-Silbigre I, Soares-Weiser K, et al. Beta-
176. Barza M, Ioannidis JPA, Cappelleri JC, et al. Single or mul- 399. Gatell JM, Ferran F, Araujo V, et al. Univariate and multi-
lactam monotherapy versus beta-lactam-aminoglycoside
tiple daily doses of aminoglycosides: a meta-analysis. BMJ. variate analyses of risk factors predisposing to auditory
combination therapy for sepsis in immunocompetent
1996;312:338-345. toxicity in patients receiving aminoglycosides. Antimicrob
patients: systemic review and meta-analysis of randomized
177. Munckhof WJ, Grayson JL, Turnidge JD. A meta-analysis Agents Chemother. 1987;31:1383-1387.
trials. BMJ. 2004;328:668.
of studies on the safety and efficacy of aminoglycosides 403. Sha S-H, Qiu JH, Schacht J. Aspirin attenuates gentamicin-
5. Paul M, Silbiger I, Rozinsky S, et al. Beta-lactam antibiotic
given either once daily or as divided doses. J Antimicrob induced hearing loss. N Engl J Med. 2006;354:1856-1857.
monotherapy versus beta-lactam-aminoglycoside antibi-
Chemother. 1996;37:645-663. 428. Vidal L, Gafter-Gvili A, Borok S, et al. Efficacy and safety
otic combination therapy for sepsis. Cochrane Database
178. Hatala R, Dinh T, Cook DJ. Once-daily aminoglycoside of aminoglycoside monotherapy: systematic review and
Syst Rev. 2009;(1):C3D003344.
dosing in immunocompetent adults: a meta-analysis. Ann meta-analysis of randomized controlled trials. J Antimicrob
6. Kumar A, Zarychanski R, Light B, et al. Early combination
Intern Med. 1996;124:717-725. Chemother. 2007;60:247-257.
antibiotic therapy yields improved survival compared with
185. Freeman CD, Strayer AH. Mega-analysis of meta-analysis: 434. Craig WA, Andes D. Aminoglycosides are useful for severe
monotherapy in septic shock: a prospensity-matched anal-
an examination of meta-analysis with an emphasis on respiratory tract infections. Semin Respir Infect. 1997;12:
ysis. Crit Care Med. 2010;38:1773-1785.
once-daily aminoglycoside comparative trials. Pharmaco- 271-277.
8. Drusano GL, Louie A. Optimization of aminoglycoside
therapy. 1996;16:1093-1102. 442. Leibovici L, Paul M, Poznanski O, et al. Monotherapy
therapy. Antimicrob Agents Chemother. 2011;55:2528-2531.
189. Rougier F, Ducher M, Maurin M, et al. Aminoglycoside versus β-lactam-aminoglycoside combination treatment
9. Leibovici L, Vidal L, Paul M. Aminoglycoside drugs in
dosages and nephrotoxicity: quantitative relationships. Clin for gram-negative bacteria: a prospective, observational
clinical practice: an evidence-based approach. J Antimicrob
Pharm. 2003;42:493-500. study. Antimicrob Agents Chemother. 1997;41:1127-1133.
Chemother. 2009;63:246-251.
207. Ryan G, Mukhopadhyay S, Singh M. Nebulized anti- 445. Martinez JA, Cobos-Triqueros N, Soriano A, et al. Influ-
21. Houghton JL, Green KD, Chen W, et al. The future of
pseudomonal antibiotics for cystic fibrosis. Cochrane Data- ence of empiric therapy with a beta-lactam alone or com-
aminoglycosides: the end or the renaissance? Chembio-
base Syst Rev. 2003;(3):CD001021. bined with an aminoglycoside on prognosis of bacteremia
chem. 2010;11:880-902.
256. Wallace AW, Jones M, Bertino JS. Evaluation of four once- due to gram-negative microorganisms. Antimicrob Agents
29. Taber HW, Muller JP, Arrow AS. Bacterial uptake of ami-
daily aminoglycoside dosing nomograms. Pharmacother- Chemother. 2010;54:3590-3596.
noglycoside antibiotics. Microbiol Rev. 1987;51:439-457.
apy. 2002;22:1077-1083. 446. Micek ST, Welch EC, Kha J, et al. Empiric combination
41. Kohanski MA, Dwyer DJ, Hayete B, et al. A common
258. Streetman DS, Nafziger AN, Destache CJ, et al. Indi­ antibiotic therapy is associated with improved outcome
mechanism of cellular death induced by bactericidal anti-
vidualized pharmacokinetic monitoring results in less against sepsis due to gram-negative bacteria: a retro­
biotics. Cell. 2007;130:797-810.
aminoglycoside-associated nephrotoxicity and fewer asso- spective analysis. Antimicrob Agents Chemother. 2010;54:
42. Kern I, Wu Y, Inocencio J, et al. Killing by bactericidal
ciated costs. Pharmacotherapy. 2001;21:443-451. 1742-1748.
antibiotics does not depend on reactive oxygen species.
260. Mingeot-Leclercq M-P, Tulkens PM. Aminoglycosides: 447. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis
Science. 2013;339:1213-1216.
nephrotoxicity. Antimicrob Agents Chemother. 1999;43: campaign: international guidelines for management of
51. Hermann T. Aminoglycoside antibiotics: old drugs and
1005-1012. severe sepsis and septic shock: 2012. Crit Care Med. 2013;
new therapeutic approaches. Cell Mol Life Sci. 2007;64:
329. Kahlmeter G, Dahlager JI. Aminoglycoside toxicity: 41:580-637.
1841-1852.
a review of clinical studies published between 1975 449. Falagas ME, Matthaiou DK, Bliziotis IA. The role of ami-
60. Magnet S, Blanchard JS. Molecular insights into amino­
and 1982. J Antimicrob Chemother. 1984;13(suppl A): noglycosides in combination with a beta-lactam for the
glycoside action and resistance. Chem Rev. 2005;105:
S9-S22. treatment of bacterial endocarditis: a meta-analysis of
477-497.
330. Lietman PS, Smith CR. Aminoglycoside nephrotoxicity in comparative trials. J Antimicrob Chemother. 2006;57:
75. Mingeot-Leclercq M-P, Glupczynski Y, Tulkins P. Amino-
humans. Rev Infect Dis. 1983;5(suppl 2):S284-S292. 639-647.
glycosides: activity and resistance. Antimicrob Agents Che-
334. Bertino JS, Booker LA, Franck PA, et al. Incidence of and 459. Safdar N, Handelsman J, Maki DG. Does combination
mother. 1999;43:727-737.
significant risk factors for aminoglycoside-associated intravenous antibiotic therapy reduce mortality in gram-
121. Craig WA. Pharmacokinetic/pharmacodynamic parame-
nephrotoxicity in patients dosed by using individualized negative bacteremia? A meta-analysis. Lancet Infect Dis.
ters: rationale for antibacterial dosing of mice and men.
pharmacokinetic monitoring. J Infect Dis. 1993;167: 2004;4:519-527.
Clin Infect Dis. 1998;26:1-10.
173-179. 465. Falagas ME, Matthaiou DK, Karveli EA, et al. Meta-
137. Moore RD, Lietman PS, Smith CR. Clinical response
340. Buchholtz K, Larsen CT, Hassager C, et al. Severity of gen- analysis: randomized controlled trials of clindamicin/
to aminoglycoside therapy: importance of the ratio of
tamicin’s nephrotoxic effect on patients with infective aminoglycoside vs β-lactam monotherapy for the treat-
peak concentration to minimal inhibitory concentrations.
endocarditis: a prospective observational cohort study of ment of intra-abdominal infections. Aliment Pharmacol
J Infect Dis. 1987;155:93-99.
373 patients. Clin Infect Dis. 2009;48:65-71. Ther. 2006;25:537-556.
139. Kashuba AD, Nafziger AN, Drusano GL, et al. Optimizing
360. Brummett RE, Fox KE. Aminoglycoside induced hearing 478. Bratzler DW, Dellinger EP, Olsen KM, et al. Clinical prac-
aminoglycoside therapy for nosocomial pneumonia caused
loss in humans. Antimicrob Agents Chemother. 1989;33: tice guidelines for antimicrobial prophylaxis in surgery. Am
by gram-negative bacteria. Antimicrob Agents Chemother.
797-800. J Health Syst Pharm. 2013;70:195-283.
1999;43:623-629.
321.e1
References 27. Ryers DH, Rando RR. Aminoglycoside binding to human
and bacterial A-site rRNA decoding region constructs.
54. Floquet C, Hatin I, Rousset JP, et al. Statistical analysis of
readthrough levels for nonsense mutations in mammalian
1. Wright GD, Berghuis AM, Mobashery S. Aminoglycoside
Bioorg Med Chem. 2001;9:2601-2608. cells reveals a major determinant of response to gentami-
antibiotics: structures, functions, and resistance. In: Rosen
28. Bryan LE, Kawan S. Roles of ribosomal binding, membrane cin. PLoS Genet. 2012;8:e1002608.
BP, Mobashery S, eds. Resolving the Antibiotic Paradox:

Chapter 25  Aminoglycosides


potential, and electron transport in bacterial uptake of 55. Hocquet D, Vogue C, El Garch F, et al. MexXY-OprM efflux
Progress in Understanding Drug Resistance and Develop-
streptomycin and gentamicin. Antimicrob Agents Che- pump is necessary for adaptive resistance of Pseudomonas
ment of New Antibiotics. New York: Plenum; 1998:27-69.
mother. 1983;23:835-845. aeruginosa to aminoglycosides. Antimicrob Agents Che-
2. Craig WA. Optimizing aminoglycoside use. Crit Care Clin.
29. Taber HW, Muller JP, Arrow AS. Bacterial uptake of ami- mother. 2003;47:1371-1375.
2011;27:107-121.
noglycoside antibiotics. Microbiol Rev. 1987;51:439-457. 56. Mao W, Warren MS, Lee A, et al. MexXY-OprM efflux
3. Vogelman B, Craig WA. Kinetics of antimicrobial activity.
30. Hancock RE, Bellido F. Antibiotic uptake: unusual results pump is required for antagonism of aminoglycosides by
J Pediatr. 1986;108:835-840.
for unusual molecules. J Antimicrob Chemother. 1992;29: divalent cation in Pseudomonas aeruginosa. Antimicrob
4. Paul M, Benuri-Silbigre I, Soares-Weiser K, et al. Beta-
235-239. Agents Chemother. 2001;45:2001-2007.
lactam monotherapy versus beta-lactam-aminoglycoside
31. Hurwitz C, Rosano CL, Landau JV. Kinetics of loss of 57. Daikos GL, Jackson GG, Lolans VT, et al. Adaptive resis-
combination therapy for sepsis in immunocompetent
viability of Escherichia coli exposed to streptomycin. J Bac- tance to aminoglycoside antibiotics from first-exposure
patients: systemic review and meta-analysis of randomized
teriol. 1962;83:1210-1216. down-regulation. J Infect Dis. 1990;162:414-420.
trials. BMJ. 2004;328:668.
32. Jackson GE, Lolans VT, Daikos GL. The inductive role of 58. Xiong Y-Q, Caillon J, Kergueris MF, et al. Adaptive resis-
5. Paul M, Silbiger I, Rozinsky S, et al. Beta-lactam antibiotic
ionic binding in the bactericidal and postexposure effects tance of Pseudomonas aeruginosa induced by aminoglyco-
monotherapy versus beta-lactam-aminoglycoside antibi-
of aminoglycoside antibiotics with implications for dosing. sides and killing kinetics in a rabbit endocarditis model.
otic combination therapy for sepsis. Cochrane Database
J Infect Dis. 1990;162:408-413. Antimicrob Agents Chemother. 1997;41:823-826.
Syst Rev. 2009;(1):C3D003344.
33. Laurent G, Kishore BK, Tulkens PM. Aminoglycoside- 59. Bush K, Miller GH. Bacterial enzymatic resistance: beta-
6. Kumar A, Zarychanski R, Light B, et al. Early combination
induced renal phospholipidosis and nephrotoxicity. lactamases and aminoglycoside-modifying enzymes. Curr
antibiotic therapy yields improved survival compared with
Biochem Pharmacol. 1990;40:2383-2392. Opin Microbiol. 1998;1:509-515.
monotherapy in septic shock: a prospensity-matched anal-
34. Bryan LE. Mechanisms of action of aminoglycoside antibi- 60. Magnet S, Blanchard JS. Molecular insights into amino­
ysis. Crit Care Med. 2010;38:1773-1785.
otics. In: Root RK, Sande MA, eds. Contemporary Issues in glycoside action and resistance. Chem Rev. 2005;105:
7. Ababneh M, Harpe S, Oinonen M, et al. Trends in amino-
Infectious Diseases. Vol 1. New Dimensions in Antimicrobial 477-497.
glycoside use and gentamicin-resistant gram-negative
Therapy. New York: Churchill Livingstone; 1984:17-36. 61. Vakulenko SB, Mobashery S. Versatility of aminoglycosides
clinical isolates in US academic medical centers: implica-
35. Hancock REW. Alterations in outer membrane permeabil- and prospects for their future. Clin Microbiol Rev. 2003;16:
tions for antimicrobial stewardship. Infect Control Hosp
ity. Annu Rev Microbiol. 1984;38:237-264. 430-450.
Epidemiol. 2012;33:594-601.
36. Peterson AA, Hancock REW, McGroarty EJ. Binding of 62. Hoffman LR, d’Argenio DA, MacCoss MJ, et al. Aminogly-
8. Drusano GL, Louie A. Optimization of aminoglycoside
polycationic antibiotics and polyamines to lipopolysaccha- coside antibiotics induce bacterial biofilm formation.
therapy. Antimicrob Agents Chemother. 2011;55:2528-2531.
rides of Pseudomonas aeruginosa. J Bacteriol. 1985;164: Nature. 2005;436:1171-1175.
9. Leibovici L, Vidal L, Paul M. Aminoglycoside drugs in
1256-1261. 63. Shurek KN, Marr AK, Taylor PK, et al. Novel genetic deter-
clinical practice: an evidence-based approach. J Antimicrob
37. Martin NL, Beveridge TJ. Gentamicin interaction with minants of low-level aminoglycoside resistance in Pseudo-
Chemother. 2009;63:246-251.
Pseudomonas aeruginosa. Antimicrob Agents Chemother. monas aeruginosa. Antimicrob Agents Chemother. 2008;52:
10. Berdy J, Aszalos A, Bostian M, et al. CRC Handbook of
1986;29:1079-1087. 4213-4219.
Antibiotic Compounds. Vol 1. Carbohydrate Antibiotics.
38. Hancock REW. Aminoglycoside uptake and mode of action 64. Krahn T, Gilmour C, Tilak J, et al. Determinants of intrin-
Boca Raton, FL: CRC Press; 1980.
with special reference to streptomycin and gentamicin. I: sic aminoglycoside resistance in Pseudomonas aeruginosa.
11. Weinstein MJ, Wagman GH, Oden EM, et al. Biological
Antagonists and mutants. J Antimicrob Chemother. 1981;8: Antimicrob Agents Chemother. 2012;56:5591-5602.
activity of the antibiotic components of the gentamicin
249-276. 65. Barclay ML, Begg EJ, Chambers ST, et al. Adaptive resis-
complex. J Bacteriol. 1967;94:789-790.
39. Hancock REW. Aminoglycoside uptake and mode of action tance to tobramycin in Pseudomonas aeruginosa lung infec-
12. Gilbert DN, Kohlhepp SJ. New sodium hydroxide digestion
with special reference to streptomycin and gentamicin. II: tion in cystic fibrosis. J Antimicrob Chemother. 1996;37:
method for measurement of renal tobramycin concentra-
Effects of aminoglycosides on cells. J Antimicrob Che- 1155-1164.
tions. Antimicrob Agents Chemother. 1986;30:361-365.
mother. 1981;8:429-445. 66. Gilleland LB, Gilleland HE, Gibson JA, et al. Adaptive resis-
13. Moellering RC Jr. In vitro antibacterial activity of the ami-
40. Nicas TI, Hancock REW. Outer membrane protein H1 of tance to aminoglycoside antibiotics in Pseudomonas aeru-
noglycoside antibiotics. Rev Infect Dis. 1983;5(suppl):
Pseudomonas aeruginosa: involvement in adaptive and ginosa. J Med Microbiol. 1984;29:41-50.
S212-S232.
mutational resistance to ethylenediamine tetraacetate, 67. Hocquet D, Vogne C, El Garch F, et al. MexXY-OprM efflux
14. Kaul M, Barbieri CM, Kerrigan JE, et al. Coupling of drug
polymyxin B, and gentamicin. J Bacteriol. 1980;143: pump is necessary for adaptive resistance of Pseudomonas
protonation to the specific binding of aminoglycosides to
872-878. aeruginosa to aminoglycosides. Antimicrob Agents Che-
the A site of 16S rRNA: elucidation of the number of drug
41. Kohanski MA, Dwyer DJ, Hayete B, et al. A common mother. 2003;47:1371-1375.
amino groups involved and their identities. J Mol Biol.
mechanism of cellular death induced by bactericidal anti- 68. Häussler SB, Tümmler B, Weissbrodt M, et al. Small-colony
2003;326:1373-1387.
biotics. Cell. 2007;130:797-810. variants of Pseudomonas aeruginosa in cystic fibrosis. Clin
15. Rocque WJ, Fesik SW, Haug A, et al. Polycation bind­
42. Kern I, Wu Y, Inocencio J, et al. Killing by bactericidal Infect Dis. 1999;29:621-625.
ing to isolated lipopolysaccharide from antibiotic-
antibiotics does not depend on reactive oxygen species. 69. Fritsche TR, Castanheira M, Miller GH, et al. Detection of
hypersusceptible mutant strains of Escherichia coli.
Science. 2013;339:1213-1216. methyltransferases conferring high-level resistance to ami-
Antimicrob Agents Chemother. 1988;32:308-313.
43. Lavitan IM, Fraimow HS, Carrasco N, et al. Tobramycin noglycosides in Enterobacteriaceae from Europe, North
16. McLaughlin JE, Reeves DS. Clinical and laboratory evi-
uptake in Escherichia coli in membrane vesicles. Antimicrob America and Latin America. Antimicrob Agents Che-
dence for inactivation of gentamicin by carbenicillin.
Agents Chemother. 1995;39:467-475. mother. 2008;52:1843-1845.
Lancet. 1971;1:261-264.
44. Kellermayer R. Translational readthrough induction of 70. Wachino J, Shibayama K, Kurokawa H. Novel plasmid-
17. Pickering LK, Rutherford I. Effect of concentration and
pathogenic nonsense mutations. Eur J Med Genet. 2006; mediated 16S rRNA m1A1408 methyltransferase, NpmA,
time upon inactivation of tobramycin, gentamicin, netilmi-
49:445-450. found in a clinically isolated Escherichia coli strain resistant
cin, mezlocillin, and piperacillin. J Pharmacol Exp Ther.
45. Bedwell DM, Kaerijak A, Benos DJ, et al. Suppression of a to structurally diverse aminoglycosides. Antimicrob Agents
1981;217:345-349.
CFTR premature stop mutation in a bronchial epithelial Chemother. 2007;51:4401-4409.
18. Walterspiel JN, Feldman S, Van R, et al. Comparative inac-
cell line. Nat Med. 1997;3:1280-1284. 71. Doi Y, Arakawa Y. 16S ribosomal RNA methylation:
tivation of isepamicin, amikacin, and gentamicin by nine
46. Clancy JP, Bebok Z, Ruiz F, et al. Evidence that systemic emerging resistance mechanism against aminoglycosides.
beta-lactams and two beta-lactamase inhibitors, cilastatin
gentamicin suppresses premature stop mutations in Clin Infect Dis. 2007;45:88-94.
and heparin. Antimicrob Agents Chemother. 1991;35:
patients with cystic fibrosis. Am J Respir Crit Care Med. 72. Yamane K, Wachino J, Doi Y, et al. Global spread of mul-
1875-1878.
2001;163:1683-1692. tiple aminoglycoside resistance genes. Emerg Infect Dis.
19. Desai NR, Shah SM, Cohen J, et al. Zosyn (piperacillin/
47. Du M, Jones JR, Lanier J, et al. Aminoglycoside suppression 2005;11:951-953.
tazobactam) reformulation: expanded compatibility and
of a premature stop mutation in a Cftr−/− mouse carrying 73. Canton R. Antibiotic resistance genes from the environ-
co-administration with lactated Ringer’s solutions and
a human CFTR-G542A transgene. J Mol Med. 2002;80: ment: a perspective through newly identified antibiotic
selected aminoglycosides. Ther Clin Risk Man. 2008;4:
595-604. resistance mechanisms in the clinical setting. Clin Micro-
303-314.
48. Howard MT, Shirts BH, Petros LM, et al. Sequence specific- biol Infect. 2009;15(suppl 1):20-25.
20. Halstenson CE, Wong MU, Herman CS, et al. Effect of
ity of aminoglycoside-induced stop codon readthrough: 74. Dozzo P, Moser HE. New aminoglycoside antibiotics.
concomitant administration of piperacillin on the disposi-
potential implication for treatment of Duchenne muscular Expert Opin Ther Pat. 2010;20:1321-1341.
tions of isepamicin and gentamicin in patients with end-
dystrophy. Ann Neurol. 2000;48:164-169. 75. Mingeot-Leclercq M-P, Glupczynski Y, Tulkins P. Amino-
stage renal disease. Antimicrob Agents Chemother. 1992;36:
49. Keeling KM, Brooks DA, Hopwood JJ, et al. Gentamicin- glycosides: activity and resistance. Antimicrob Agents Che-
1832-1836.
mediated suppression of Hurler syndrome stop mutations mother. 1999;43:727-737.
21. Houghton JL, Green KD, Chen W, et al. The future of
restores a low level of a-2-iduronidase activity and restores 76. Fong DH, Berghuis AM. Substrate promiscuity of an
aminoglycosides: the end or the renaissance? Chembio-
lysosomal glycosaminoglycan accumulation. Hum Mol aminoglycoside antibiotic resistance enzyme via target
chem. 2010;11:880-902.
Genet. 2001;10:291-299. mimicry. EMBO J. 2002;21:2323-2331.
22. Walter F, Vicens Q, Westhof E. Aminoglycoside-RNA
50. Schulz A, Sangkuhl K, Lennert T, et al. Aminoglycoside 77. Shaw KJ, Rather PN, Hare RS. Molecular genetics of ami-
interactions. Curr Opin Chem Biol. 1999;3:694-704.
pretreatment partially restores the function of truncated noglycoside resistance genes and familial relationships of
23. Lynch SR, Recht MI, Puglisi JD. Biochemical and nuclear
V2 vasopressin receptors found in patients with nephro- the aminoglycoside-modifying enzymes. Microbiol Rev.
magnetic resonance studies of aminoglycoside-RNA com-
genic diabetes insipidus. J Clin Endocrinol Metab. 2002; 1993;57:138-163.
plexes. Methods Enzymol. 2000;317:240-261.
87:5247-5257. 78. Vakulenko SB, Donabedian SM, Voskresensky AM, et al.
24. Vicens Q, Westhoff E. RNA as a drug target: the case of
51. Hermann T. Aminoglycoside antibiotics: old drugs and Multiplex PCR for detection of aminoglycoside resistance
aminoglycosides. Chembiochem. 2003;4:1018-1023.
new therapeutic approaches. Cell Mol Life Sci. 2007;64: genes in enterococci. Antimicrob Agents Chemother. 2003;
25. Lynch SR, Puglisi JD. Structural origins of aminoglycoside
1841-1852. 47:1423-1426.
specificity for prokaryotic ribosomes. J Mol Biol. 2001;306:
52. Linde L, Boelz S, Nissim-Rafinia M, et al. Nonsense- 79. Zhang W, Fisher JF, Mobashery S. The bifunctional
1037-1058.
mediated mRNA decay affects nonsense transcript levels enzymes of antibiotic resistance. Curr Opin Microbiol.
26. Francois B, Russell RJM, Murray JB, et al. Crystal struc-
and governs response of cystic fibrosis patients to gentami- 2009;12:505-511.
tures of complexes between aminoglycosides and decoding
cin. J Clin Invest. 2007;117:683-692. 80. Moellering RC. The enterococcus: a classic example of the
A site oligonucleotides: role of the number of rings and
53. Rowe SM, Clancy JP. Pharmaceuticals targeting nonsense impact of antimicrobial resistance on therapeutic options.
positive charges in the specific binding leading to miscod-
mutations in genetic diseases. Biodrugs. 2009;23:165-174. J Antimicrob Chemother. 1991;28:1-12.
ing. Nucleic Acids Res. 2005;33:5677-5690.
321.e2
81. Chow JW. Aminoglycoside resistance in enterococci. Clin 106. Barmada S, Kohlhepp S, Leggett J, et al. Correlation of factor: comparison of the pathogenic potential of encapsu-
Infect Dis. 2000;31:586-589. tobramycin-induced inhibition of protein synthesis with lated and unencapsulated strains. J Infect Dis. 1977;136:
82. Aslangul E, Ruimy R, Chau F, et al. Relationship between postantibiotic effect in Escherichia coli. Antimicrob Agents 82-89.
the level of acquired resistance to gentamicin and syner- Chemother. 1993;27:2678-2683. 133. Nichols RL, Smith JW. Wound and intra-abdominal infec-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

gism with amoxicillin in Enterococcus faecalis. Antimicrob 107. Craig WA, Ebert SC. Killing and regrowth of bacteria in tions: microbiological considerations and approaches to
Agents Chemother. 2005;49:4144-4148. vitro: a review. Scand J Infect Dis Suppl. 1991;74:63-70. treatment. Clin Infect Dis. 1993;16(suppl 4):S266-S277.
83. Costa Y, Galiman M, Leclercq R, et al. Characterization 108. Hessen MT, Pitsakis PG, Levison ME. Absence of a post- 134. Vandaux P, Waldvogel FA. Gentamicin inactivation in
of the chromosomal aac(6′)-Ii gene specific for Entero­ antibiotic effect in experimental Pseudomonas endocarditis purulent exudates: role of cell lysis. J Infect Dis. 1980;142:
coccus faecium. Antimicrob Agents Chemother. 1993;37: treated with imipenem, with or without gentamicin. J Infect 586-593.
1896-1903. Dis. 1988;158:542-548. 135. Kapusnik JE, Hackbarth CJ, Chambers HF, et al. Single,
84. Karlowsky JA, Jones ME, Thornsberry CT, et al. Trends in 109. Gudmundsson S, Einarsson S, Erlendstotter H, et al. The large daily dosing vs intermittent dosing of tobramycin for
antimicrobial susceptibilities among Enterobacteriaceae postantibiotic effect of antimicrobial combinations in a treating experimental Pseudomonas aeruginosa pneumo-
isolated from hospitalized patients in the United States neutropenic murine thigh infection model. J Antimicrob nia. J Infect Dis. 1988;158:7-12.
from 1998 to 2001. Antimicrob Agents Chemother. 2003; Chemother. 1993;31(suppl D):S177-S191. 136. Gilbert DN. Once-daily aminoglycoside therapy. Antimi-
47:1672-1680. 110. Hunter TH. Use of streptomycin in the treatment of bacte- crob Agents Chemother. 1991;35:339-405.
85. Karlowsky JA, Draghi DC, Jones ME, et al. Surveillance for rial endocarditis. Am J Med. 1947;2:436-442. 137. Moore RD, Lietman PS, Smith CR. Clinical response to
antimicrobial susceptibility among clinical isolates of Pseu- 111. Craig WA. Post-antibiotic effects in experimental infection aminoglycoside therapy: importance of the ratio of peak
domonas aeruginosa and Acinetobacter baumannii from models: relationship to in vitro phenomena and to treat- concentration to minimal inhibitory concentrations.
hospitalized patients in the United States, 1998 to 2001. ment of infections in man. J Antimicrob Chemother. J Infect Dis. 1987;155:93-99.
Antimicrob Agents Chemother. 2003;47:1681-1688. 1993;31(suppl D):S149-S158. 138. Mouton JW, van Ogtrop ML, Andes D, et al. Use of phar-
86. Diekema DJ, Pfaller MA, Jones RN, et al. Survey of blood- 112. Drusano GL. Role of pharmacokinetics in the outcome macodynamic indices to predict efficacy of combination
stream infections due to gram-negative bacilli: frequency of infections. Antimicrob Agents Chemother. 1988;32: therapy in vivo. Antimicrob Agents Chemother. 1999;43:
of occurrence and antimicrobial susceptibility of isolates 289-297. 2473-2478.
collected in the United States, Canada, and Latin America 113. Holm SE. Interaction between beta-lactam and other anti- 139. Kashuba AD, Nafziger AN, Drusano GL, et al. Optimizing
for the SENTRY Antimicrobial Surveillance program, biotics. Rev Infect Dis. 1986;8(suppl 3):S305-S314. aminoglycoside therapy for nosocomial pneumonia caused
1997. Clin Infect Dis. 1999;29:595-607. 114. Bowers DR, Liew YX, Lye DC, et al. Outcome of appro­ by gram-negative bacteria. Antimicrob Agents Chemother.
87. Miller GH, Sabatelli FJ, Hare RS, et al. The most frequent priate empiric combination versus monotherapy for 1999;43:623-629.
aminoglycoside resistance mechanisms—changes with Pseudomonas aeruginosa bacteremia. Antimicrob Agents 140. Smith PF, Ballow CH, Booker BM, et al. Pharmacokinetics
time and geographic area: a reflection of aminoglycoside Chemother. 2013;57:1270-1274. and pharmacodynamics of aztreonam and tobramycin in
usage patterns? Aminoglycoside Resistance Study Groups. 115. Chen CH, Gau V, Zhang DD, et al. Statistical metamodel- hospitalized patients. Clin Ther. 2001;23:1231-1244.
Clin Infect Dis. 1997;24(suppl 1):S46-S62. ing for revealing synergistic antimicrobial interactions. 141. Drusano GL, Ambrose PG, Bhavnani SM, et al. Back to the
88. Chow JW, Kak V, You I, et al. The changing nature of PLoS One. 2010;5:e15472. future: using aminoglycosides again and how to dose them
aminoglycoside resistance mechanisms and prevalence of 116. Andriole VT. Aminoglycoside antibiotics: antibacterial optimally. Clin Infect Dis. 2007;45:753-760.
newly recognized resistance mechanisms in Turkey. Clin efficacy in animal models of infection. J Infect Dis. 1983; 142. Tamma PD, Cosgrove SE, Maragakis LL. Combination
Microbiol Infect. 2001;7:470-478. 5(suppl 2):S233-S249. therapy for treatment of infections with gram-negative
89. Vanhoof R, Nyseen HJ, Bossuyt EV, et al. Aminoglycoside 117. Fantin B, Carbon C. In vivo antibiotic synergism: contribu- bacteria. Clin Microbiol Rev. 2012;25:450-470.
resistance in gram-negative blood isolates from various tion of animal models. Antimicrob Agents Chemother. 1992; 143. Klastersky J, Prevost JM, Meunier-Carpentier F, et al. Com-
hospitals in Belgium and the Grand Duchy of Luxembourg. 36:907-912. parative trials of single dose vs twice daily sisomicin in
J Antimicrob Chemother. 1999;44:483-488. 118. Chavanet P, Collin F, Muggeo E, et al. The in vivo activity bacteriuric patients. J Clin Pharmacol. 1997;17:520-528.
90. Schwaber MJ, Carmeli Y. Carbapenem-resistant Entero- of co-amoxiclav with netilmicin against experimental 144. Nordstrom L, Ringberg H, Cronberg S, et al. Does admin-
bacteriaceae: a potential threat. JAMA. 2008;300: methicillin and gentamicin resistant Staphylococcus epider- istration of an aminoglycoside in single daily dose affect its
2911-2913. midis infection in rabbits. J Antimicrob Chemother. 1993; efficacy and toxicity? J Antimicrob Chemother. 1990;25:
91. Guimaraes MA, Sage R, Noone P. The comparative activity 31:129-138. 159-173.
of aminocyclitol antibiotics against 773 aerobic gram- 119. Trautman M, Bruckner O, Marre R, et al. Comparative 145. Hansen M, Achen F, Carstensen C, et al. Once-versus
negative rods and staphylococci isolated from hospitalized efficacy of ciprofloxacin, ceftazidime and gentamicin, given thrice-daily dosing of netilmicin in febrile immuno­
patients. J Antimicrob Chemother. 1985;16:555-562. alone or in combination, in a model of experimental sep- compromised patients: a randomized, controlled study
92. Gerding DN, Larson TA. Aminoglycoside resistance in ticemia due to Klebsiella pneumoniae in neutropenic mice. of efficacy and safety. J Drug Dev. 1988;1(suppl 3):
gram-negative bacilli during increased amikacin use. Am J Infection. 1988;16:49-53. S119-S124.
Med. 1985;79:1-7. 120. Chadwick EK, Shulman ST, Yogev R. Correlation of anti- 146. Muijsken MA, Vreede RW, Van Dijk WC, et al. A random-
93. Bengtsson S, Bernarder S, Brorson JE, et al. In vitro ami- biotic synergy in vitro and in vivo: use of an animal model ized clinical study of efficacy and safety of once daily versus
noglycoside resistance of gram-negative bacilli and staphy- of neutropenic gram-negative sepsis. J Infect Dis. 1986;154: conventional dosing of netilmicin in patients with severe
lococci isolated from blood in Sweden 1980-1984. Scand J 670-675. infections. J Drug Dev. 1988;1(suppl 3):S145-S146.
Infect Dis. 1986;18:257. 121. Craig WA. Pharmacokinetic/pharmacodynamic parame- 147. Tulkens PM, Clerckx-Braun F, Donnez J, et al. Safety and
94. Maurin M, Raoult D. Use of aminoglycosides in treatment ters: rationale for antibacterial dosing of mice and men. efficacy of aminoglycosides once-a-day: experimental data
of infections due to intracellular bacteria. Antimicrob Clin Infect Dis. 1998;26:1-10. and randomized, controlled evaluation in patients suffer-
Agents Chemother. 2001;45:2977-2986. 122. Vogelman B, Gudmundsson S, Leggett J, et al. Correlation ing from pelvic inflammatory disease. J Drug Dev. 1988;
95. Gilbert DN, Moellering RC, Eliopoulos GM, et al. Sanford of antimicrobial pharmacokinetic parameters with thera- 1(suppl 3):S71-S83.
Guide to Antimicrobial Therapy. 42nd ed. Sperryville, VA: peutic efficacy in an animal model. J Infect Dis. 1988;158: 148. Hollender LF, Bahnini J, DeManzini N, et al. A multicentric
Antimicrobial Therapy; 2012. 831-847. study of netilmicin once daily versus thrice daily in patients
96. Rolain J-M, Maurin M, Raoult D. Bactericidal effect of 123. Graham JC, Gould FK. Role of aminoglycosides in the with appendicitis and other intra-abdominal infections.
antibiotics on Bartonella and Brucella spp: clinical implica- treatment of bacterial endocarditis. J Antimicrob Che- J Antimicrob Chemother. 1989;23:773-783.
tions. J Antimicrob Chemother. 2000;46:811-814. mother. 2002;49:437-444. 149. Mauracher EH, Lau WY, Kartowisastro H, et al. Compari-
97. Salah AB, Messaoud NB, Guedri E, et al. Topical paromo- 124. Gavalda J, Onrubia PL, Gomez MT, et al. Efficacy of ampi- son of once-daily and thrice-daily netilmicin regimens in
mycin with or without gentamicin for cutaneous leish- cillin combined with ceftriaxone and gentamicin in the serious system infections: a multicenter study in six Asian
maniasis. N Engl J Med. 2013;368:524-532. treatment of experimental endocarditis due to Enterococcus countries. Clin Ther. 1989;11:604-613.
98. Peddie BA, Chambers ST. Effects of betaines and urine on faecalis with no high-level resistance to aminoglycosides. 150. Strum AW. Netilmicin in the treatment of gram-negative
the antibacterial activity of aminoglycosides. J Antimicrob J Antimicrob Chemother. 2003;52:514-517. bacteremia: single daily versus multiple daily dosage.
Chemother. 1993;31:481-488. 125. Fantin B, Pangon B, Potel G, et al. Ceftriaxone-netilmicin J Infect Dis. 1989;159:931-937.
99. Eliopoulos GM, Moellering RC. Antimicrobial combina- combination in single-daily-dose treatment of experimen- 151. DeVries PJ, Verkooyen RP, Leguit P, et al. Prospective ran-
tions. In: Lorian V, ed. Antibiotics in Laboratory Medicine. tal Escherichia coli endocarditis. Antimicrob Agents Che- domized study of once-daily versus thrice-daily netilmicin
4th ed. Baltimore: Williams & Wilkins; 1996:330-383. mother. 1989;33:767-770. regimens in patients with intra-abdominal infections. Eur
100. Dudley MN, Zinner SH. Single daily dosing of amikacin 126. Michea-Hamzehpour M, Auckenthaler R, Regamey P, et al. J Clin Microbiol Infect Dis. 1990;9:161-168.
in an in vitro model. J Antimicrob Chemother. 1991;27 Resistance occurring after fluoroquinolone therapy of 152. Ter Braak EW, deVried PJ, Bouter KP, et al. Once-daily
(suppl C):15-19. experimental Pseudomonas aeruginosa peritonitis. Antimi- dosing regimen for aminoglycoside plus beta-lactam com-
101. Blaser J, Stone BB, Zinner SH. Efficacy of intermittent crob Agents Chemother. 1987;31:1803-1808. bination therapy of serious bacterial infections: compara-
versus continuous administration of netilmicin in a two- 127. Michea-Hamzehpour M, Pechere JC, Marchou B, et al. tive trial with netilmicin plus ceftriaxone. Am J Med. 1990;
compartment in vitro model. Antimicrob Agents Che- Combination therapy: a way to limit emergence of resis- 89:58-66.
mother. 1985;27:343-349. tance? Am J Med. 1986;80(suppl 6B):S138-S142. 153. Giamarellou H, Yiallouros K, Petrikkos G, et al. Compara-
102. Blaser J, Stone BB, Groner MC, et al. Comparative study 128. Pechere JC, Marchou B, Michea-Hamzehpour M, et al. tive kinetics and efficacy of amikacin administered once or
with enoxacin and netilmicin in a pharmacodynamic Emergence of resistance after therapy with antibiotics used twice daily in the treatment of systemic gram-negative
model to determine importance of ratio of antibiotic peak alone or combined in a murine model. J Antimicrob Che- infections. J Antimicrob Chemother. 1991;27(suppl C):
concentration to MIC for bactericidal activity and emer- mother. 1986;17(suppl A):S11-S18. S73-S79.
gence of resistance. Antimicrob Agents Chemother. 1987;31: 129. Vastola AP, Brandel J, Craig WA. Selection of 154. Marik PE, Lipman J, Kobilski S, et al. A prospective ran-
1054-1060. aminoglycoside-resistant variants of Pseudomonas aerugi- domized study comparing once-versus twice-daily amika-
103. Vogelman B, Gudmundsson S, Turnidge J, et al. In vivo nosa in an in vivo model. J Infect Dis. 1982;146:691-697. cin dosing in critically ill adult and pediatric patients.
postantibiotic effect in a thigh infection in neutropenic 130. Weinstein WM, Onderdonk AB, Bartlett JG, et al. Anti­ J Antimicrob Chemother. 1991;28:753-764.
mice. J Infect Dis. 1988;157:287-298. microbial therapy of experimental intra-abdominal sepsis. 155. Van de Auwera P, Meunier J, Ibrahim S, et al. Pharmaco-
104. Craig WA, Gudmundsson S. Postantibiotic effect. In: J Infect Dis. 1975;132:282-286. dynamic and toxicity of netilmicin (5 mg/kg/day) given
Lorain V, ed. Antibiotics in the Laboratory. 4th ed. Balti- 131. Weinstein WM, Onderdonk AB, Bartlett JG, et al. once daily or in three divided doses to cancer patients with
more: Williams & Wilkins; 1996:296-329. Experimental intra-abdominal abscesses in rats: develop- urinary tract infections. Antimicrob Agents Chemother.
105. Fantin B, Ebert S, Leggett J, et al. Factors affecting duration ment of an experimental model. Infect Immun. 1974;10: 1991;35:640-647.
of in vivo postantibiotic effect for aminoglycosides against 1250-1255. 156. Vigano A, Prinicipi N, Brivio L, et al. Comparison of
gram-negative bacilli. J Antimicrob Chemother. 1990;27: 132. Onderdonk AB, Kasper DL, Cisneros RL, et al. The capsu- 5 mg of netilmicin per kilogram of body weight once
829-836. lar polysaccharide of Bacteroides fragilis as a virulence daily versus 2 mg per kilogram thrice daily for treatment
321.e3
of gram-negative pyelonephritis in children. Antimicrob 2 mg per kilogram thrice for treatment of gram-negative of four antibiotics. Antimicrob Agents Chemother. 1982;21:
Agents Chemother. 1992;36:1499-1503. pyelonephritis in children. Antimicrob Agents Chemother. 873-880.
157. Gonzalez P, Aguado JM, Martin MA, et al. Once-daily ami- 1992;36:1499-1503. 211. Zaske DE, Strate RG, Kohls PR. Amikacin pharmacokinet-
noglycoside dosing [letter]. Lancet. 1993;341:895. 182. Nicolau DP, Freeman CD, Bellineau PP, et al. Experience ics: wide interpatient variation in 98 patients. J Clin Phar-

Chapter 25  Aminoglycosides


158. Maller R, Ahrne H, Holmen C, et al. Once-versus twice- with a once-daily aminoglycoside program administered to macol. 1991;31:158-163.
daily amikacin regimen: efficacy and safety in systemic 2184 adult patients. Antimicrob Agents Chemother. 1995; 212. Bailey DN, Briggs JR. Gentamicin and tobramycin binding
gram-negative infections. J Antimicrob Chemother. 1993; 39:650-655. to human serum in vitro. J Anal Toxicol. 2004;28:187-189.
31:939-948. 183. Freeman CD, Nicolau DP, Belliveau PP, et al. Once-daily 213. Hickling KG, Begg EJ, Perry RE, et al. Serum aminoglyco-
159. Vanhaeverbeek M, Siska G, Douchamps J, et al. Compari- dosing of aminoglycosides: review and recommendations side clearance is predicted as poorly by renal aminoglyco-
son of the efficacy and safety of amikacin once or twice-a- for clinical practice. J Antimicrob Chemother. 1997;39: side clearance as by creatinine clearance in critically ill
day in the treatment of severe gram-negative infections in 677-686. patients. Crit Care Med. 1991;19:1041-1047.
the elderly. Int J Clin Pharmacol Ther Toxicol. 1993;31: 184. Gilbert DN. Meta-analyses are no longer required for 214. Schwartz SN, Pazin GJ, Lyon JA, et al. A controlled inves-
153-156. determining the efficacy of single daily dosing of amino- tigation of the pharmacokinetics of gentamicin and tobra-
160. Gibson J, Johnson L, Snowson L, et al. Single daily ceftri- glycosides [editorial response]. Clin Infect Dis. 1997;24: mycin in obese subjects. J Infect Dis. 1978;138:499-505.
axone and tobramycin in the empirical management of 816-819. 215. Kunar MJ, Mak LL, Lietman PS. Localization of
febrile neutropenic patients: a randomized trial. Int J 185. Freeman CD, Strayer AH. Mega-analysis of meta-analysis: 3
H-gentamicin in the proximal renal tubule of the mouse.
Hematol. 1993;58:63-72. an examination of meta-analysis with an emphasis on Antimicrob Agents Chemother. 1979;15:131-133.
161. Mendes da Costa P, Kaufman L. Amikacin once daily plus once-daily aminoglycoside comparative trials. Pharmaco- 216. Rodriguez V, Stewart D, Bodey GP. Gentamicin sulfate
metronidazole versus amikacin twice daily plus metroni- therapy. 1996;16:1093-1102. distribution in body fluids. Clin Pharmacol Ther. 1970;11:
dazole in colorectal surgery. Hepatogastroenterology. 1992; 186. Freeman CD, Strayer AH. Mega-analysis of meta-analysis: 275-281.
39:350-354. an examination of meta-analysis with an emphasis on 217. Dee TH, Kozin F. Gentamicin and tobramycin penetration
162. Fan ST, Lau WY, Teoh-Chan CH, et al. Once daily admin- once-daily aminoglycoside comparative trials. Pharmaco- into synovial fluid. Antimicrob Agents Chemother. 1977;12:
istration of netilmicin compared with thrice daily, both in therapy. 1996;16:1093-1102. 548-549.
combination with metronidazole, in gangrenous and per- 187. Kale-Pradhan PB, Habowski SR, Chase HC, et al. Once- 218. Panidis D, Markantonis SL, Boutzouka E, et al. Penetration
forated appendicitis. J Antimicrob Chemother. 1988;22: daily aminoglycosides: a meta-analysis of nonneutropenic of gentamicin into the alveolar lining fluid of critically ill
69-74. and neutropenic adults. J Pharm Technol. 1998;14:22-29. patients with ventilator-associated pneumonia. Chest.
163. Prins JM, Buller HP, Kuijper EJ, et al. Once versus thrice 188. Conil JM, Georges B, Ruiz S, et al. Tobramycin disposition 2005;128:545-552.
daily gentamicin in patients with serious infection. Lancet. in ICU patients receiving a once-daily regimen: population 219. Carcas AJ, Garcia-Satue JL, Zapater P, et al. Tobramycin
1993;341:335-339. approach and dosage simulations. Brit J Clin Pharmacol. penetration into epithelial lining fluid of patients with
164. Raz R, Adawi M, Romano S. Intravenous administration of 2010;71:61-71. pneumonia. Clin Pharmacol Ther. 1999;65:245-250.
gentamicin once daily versus thrice daily in adults. Eur J 189. Rougier F, Ducher M, Maurin M, et al. Aminoglycoside 220. Pitt HA, Roberts RB, Johnson WD Jr. Gentamicin levels in
Clin Microb Infect Dis. 1995;14:88-91. dosages and nephrotoxicity: quantitative relationships. Clin the human biliary tract. J Infect Dis. 1973;127:299-302.
165. Rozdzinsk E, Kern WV, Reichle A, et al. Once-daily versus Pharm. 2003;42:493-500. 221. Reckziegel R, Maguilnik I, Goldami LZ. Gentamicin con-
thrice-daily dosing of netilmicin in combination with beta- 190. Rybak MJ, Abate BJ, Kang SL, et al. Prospective evaluation centration in bile after once-daily versus thrice-daily
lactam antibiotics as empirical therapy for febrile neutro- of the effect of an aminoglycoside dosing regimen on rates dosing of 4 mg/kg/day. J Antimicrob Chemother. 2001;48:
penic patients. J Antimicrob Chemother. 1993;31:585-598. of observed nephrotoxicity and ototoxicity. Antimicrob 327-329.
166. Ibrahim S, Derde MP, Kaufman L, et al. Safety, pharmaco- Agents Chemother. 1999;43:1549-1555. 222. Bernard B, Garcia-Gazases SJ, Ballard CA, et al. Tobramy-
kinetics and efficacy of once-a-day netilmicin and amika- 191. Hansen M, Christrup LL, Jarlov JO, et al. Gentamicin cin maternal-fetal pharmacology. Antimicrob Agents Che-
cin versus their conventional schedules in patients suffering dosing in critically ill patients. Acta Anesthesiol Scand. mother. 1977;11:688-694.
from pelvic inflammatory disease. Ren Fail. 1990;12: 2001;45:734-740. 223. Rahal JJ Jr, Hyams PJ, Simberkoff MS, et al. Combined
199-203. 192. Kim MJ, Bertino JS Jr, Erb TA, et al. Application of Bayes intrathecal and intramuscular gentamicin for gram-
167. International Antimicrobial Therapy Cooperative Group theorem to aminoglycoside-associated nephrotoxocity: negative meningitis. N Engl J Med. 1974;290:1394-1398.
of the European Organization for Research and Treatment comparison of extended-interval dosing, individualized 224. Kaiser AB, McGee ZA. Aminoglycoside therapy of gram-
of Cancer. Efficacy and toxicity of single daily doses of pharmacokinetic monitoring, and multiple-daily dosing. negative bacillary meningitis. N Engl J Med. 1975;293:
amikacin and ceftriaxone versus multiple daily doses of J Clin Pharmacol. 2004;44:1072-1074. 1215-1220.
amikacin and ceftazidime for infection in patients with 193. Beauchamp D, Labrecque G. Chronobiology and chrono- 225. Wirt TC, McGee ZA, Oldfield EH, et al. Intraventricular
cancer and granulocytopenia. Ann Intern Med. 1993;199: toxicology of antibiotics and aminoglycosides. Adv Drug administration of amikacin for complicated gram-negative
584-593. Deliv Rev. 2007;59:896-903. meningitis and ventriculitis. J Neurosurg. 1979;50:95-99.
168. Maller R, Isaksson B, Nilsson L, et al. A study of amikacin 194. Lowenthal MR, Dobson PM. Tobramycin and gentamicin 226. Cracken GH Jr, Mize S, Threlkeld N. Intraventricular gen-
given once versus twice daily in serious infections. J Anti- can safely be given by slow push. J Antimicrob Chemother. tamicin therapy in gram-negative bacillary meningitis of
microb Chemother. 1998;22:75-79. 2010;65:2049-2050. infancy. Lancet. 1980;1:787-791.
169. Blaser J, Simmen HP, Thrunheer U, et al. Nephrotoxicity, 195. Barza M, Lauermann M. Why monitor serum levels of 227. Baum J. Infections of the eye. Clin Infect Dis. 1995;21:
high frequency ototoxicity, efficacy and serum kinetics of gentamicin? Clin Pharmacokinet. 1978;3:202-215. 479-488.
once versus thrice daily dosing of netilmicin in patients 196. Kunin CM, Chalmers TC, Leevy CM, et al. Absorption of 228. Barza M, Kane A, Baum J. Comparison of the effects of
with serious infections. J Antimicrob Chemother. 1995;36: orally administered neomycin and kanamycin. N Engl J continuous and intermittent systemic administration on
803-814. Med. 1960;262:380-385. the penetration of gentamicin into infected rabbit eyes.
170. Koo J, Tight R, Rajkumar V, et al. Comparison of once- 197. Breen KJ, Bryant RE, Levinson JD, et al. Neomycin absorp- J Infect Dis. 1983;147:144-148.
daily versus pharmacokinetic dosing of aminoglycosides in tion in man. Ann Intern Med. 1972;76:211-218. 229. Gorden TB, Cunningham RD. Tobramycin levels in
elderly patients. Am J Med. 1996;101:177-183. 198. Bamford MFM, Jones LF. Deafness and biochemical imbal- aqueous humor after subconjunctival injection in humans.
171. Elhanan K, Siplovich L, Ra R. Gentamicin once-daily ance after burns treatment with topical antibiotics in young Am J Ophthalmol. 1982;93:107-110.
versus thrice-daily in children. J Antimicrob Chemother. children. Arch Dis Child. 1978;53:326-329. 230. Rubenstein E, Goldfarb J, Keren G, et al. The penetration
1995;35:327-332. 199. Pittinger CB, Adamson R. Antibiotic blockade of neuro- of gentamicin into the vitreous humor in man. Invest Oph-
172. Gilbert DN, Lee BL, Dworkin RJ, et al. A randomized com- muscular function. Annu Rev Pharmacol. 1972;12:169-184. thalmol Vis Sci. 1983;24:637-639.
parison of the safety and efficacy of once-daily gentamicin 200. Chamberlain G, Needham P. The absorption of antibiotics 231. Wood MJ, Farrell W. Comparison of urinary excretion of
or thrice-daily gentamicin in combination with ticarcillin- from the bladder. J Urol. 1976;116:172-173. tobramycin and gentamicin in adults. J Infect Dis. 1976;
clavulanate. Am J Med. 1998;105:182-191. 201. Lifschitz MI, Denning CR. Safety of kanamycin aerosol. 134(suppl):S133-S136.
173. Bailey TC, Little JR, Littenberg B, et al. A meta-analysis of Clin Pharmacol Ther. 1971;12:91-95. 232. Kahlmeter G, Kamme G. Prolonged excretion of gentami-
extended interval dosing versus multiple daily dosing of 202. Odio W, VanLeier E, Klastersky J. Concentrations of gen- cin in a patient with unimpaired renal function. Lancet.
aminoglycosides. Clin Infect Dis. 1997;24:786-795. tamicin in bronchial secretions after intramuscular and 1975;1:286.
174. Ali MZ, Goetz B. Meta-analysis of the relative efficacy and endotracheal administration. J Clin Pharmacol. 1975;15: 233. Kahlmeter G. Netilmicin: clinical pharmacokinetics and
toxicity of once-daily versus multiple daily dosing of ami- 518-524. aspects of dosage schedules: an overview. Scand J Infect Dis.
noglycosides. Clin Infect Dis. 1997;24:796-809. 203. Geller DE, Pitlick WH, Nardella PA, et al. Pharmacokinet- 1980;23:74-81.
175. Hatala R, Dinh TT, Cook DJ. Once-daily aminoglycoside ics and bioavailability of aerosolized tobramycin in cystic 234. Laskin OL, Longstreth JA, Smith CR, et al. Netilmicin and
dosing for immunocompromised adults: a systematic fibrosis. Chest. 2002;122:219-226. gentamicin multidose kinetics in normal subjects. Clin
review. Clin Infect Dis. 1997;24:810-815. 204. Ramsey BW, Dorkin HL, Eisenberg JD, et al. Efficacy of Pharmacol Ther. 1983;34:644-650.
176. Barza M, Ioannidis JPA, Cappelleri JC, et al. Single or mul- aerosolized tobramycin in patients with cystic fibrosis. 235. Wilson TW, Mahon WA, Inaba T, et al. Elimination of triti-
tiple daily doses of aminoglycosides: a meta-analysis. BMJ. N Engl J Med. 1993;328:1740-1746. ated gentamicin in normal human subjects and in patients
1996;312:338-345. 205. Ramsey BW, Dorkin HL, Eisenberg JD, et al. Efficacy of with severely impaired renal function. Clin Pharmacol
177. Munckhof WJ, Grayson JL, Turnidge JD. A meta-analysis aerosolized tobramycin in patients with cystic fibrosis. Ther. 1973;14:815-822.
of studies on the safety and efficacy of aminoglycosides N Engl J Med. 1993;328:1740-1746. 236. Pea F, Viale P, Pavan F, et al. Pharmacokinetic consider-
given either once daily or as divided doses. J Antimicrob 206. Prescribing information for tobramycin solution for inha- ations for antimicrobial therapy in patients receiving renal
Chemother. 1996;37:645-663. lation (package insert). Seattle, WA: Pathogenesis Corpora- replacement therapy. Clin Pharm. 2007;46:997-1038.
178. Hatala R, Dinh T, Cook DJ. Once-daily aminoglycoside tion; January 1998. 237. Fabre J, Rudhardt M, Blanchard P, et al. Persistence of siso-
dosing in immunocompetent adults: a meta-analysis. Ann 207. Ryan G, Mukhopadhyay S, Singh M. Nebulized anti- micin and gentamicin in renal cortex and medulla com-
Intern Med. 1996;124:717-725. pseudomonal antibiotics for cystic fibrosis. Cochrane Data- pared with other organs and serum of rats. Kidney Int.
179. Galloe AM, Gaudal N, Christensen HR, et al. Aminogly- base Syst Rev. 2003;(3):CD001021. 1976;10:444-449.
cosides: single or multiple daily dosing—a meta-analysis 208. Touw DJ, Jacobs FAH, Brimicombe RW, et al. Pharmaco- 238. Schentag JJ, Jusko WJ. Renal clearance and tissue accumu-
on efficacy and safety. Eur J Clin Pharmacol. 1995;48: kinetics of aerosolized tobramycin in adult patients with lation of gentamicin. Clin Pharmacol Ther. 1977;22:
39-43. cystic fibrosis. Antimicrob Agents Chemother. 1997;41: 364-370.
180. Ferriols-Lisart R, Alos-Alminana M. Effectiveness and 184-187. 239. Laskin OL, Longstreth JA, Smith CR, et al. Netilmicin and
safety of once-daily aminoglycosides: a meta-analysis. Am 209. Barza M, Brown RB, Shen D, et al. Predictability of blood gentamicin multidose kinetics in normal subjects. Clin
J Health Syst Pharm. 1996;53:1141-1150. levels of gentamicin in man. J Infect Dis. 1975;132:165-174. Pharmacol Ther. 1983;34:644-650.
181. Vigano A, Principi N, Brivio L, et al. Comparison of 5 mg 210. Van Etta LL, Kravitz GR, Russ TE, et al. Effect of 240. Schentag JJ, Jusko WJ. Gentamicin persistence in the body.
of netilmicin per kilogram of body weight once daily versus method of administration on extravascular penetration Lancet. 1977;1:485.
321.e4
241. Demczar DJ, Nafziger AN, Bartino JS Jr. Pharmacokinetics isolated from rat kidney but not from rat brain. J Antimi- 296. Peterson LN, Borzecki JS. Inhibition of tobramycin reab-
of gentamicin at traditional versus high doses: implications crob Chemother. 1985;15:715-721. sorption in nephron segments by metabolic alkalosis.
for once-daily aminoglycoside dosing. Antimicrob Agents 269. Loveless MO, Kohlhepp SJ, Gilbert DN. The influence of Kidney Int. 1990;37:1492-1499.
Chemother. 1997;41:1115-1159. aminoglycoside antibiotics on the in vitro function of rat 297. Chiu PJS, Miller GH, Long JF, et al. Renal uptake and
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

242. Cockcroft DW, Gault MH. Prediction of creatinine clear- liver ribosomes. J Lab Clin Med. 1984;103:294-303. nephrotoxicity of gentamicin during urinary alkalinization
ance from serum creatinine. Nephron. 1976;16:31-41. 270. Sastrasink M, Weinberg JM, Humes HD. The effect of gen- in rats. Clin Exp Pharmacol Physiol. 1979;6:317-326.
243. Reichley RM, Ritchie DJ, Bailey TC. Analysis of various tamicin on calcium uptake by renal mitochondria. Life Sci. 298. Thompson JR, Simonsen R, Spindler MA, et al. Protective
creatinine clearance formulas in predicting gentamicin 1982;30:2309-2315. effect of KCl loading in gentamicin nephrotoxicity. Am J
elimination in patients with low serum creatinine. Pharma- 271. Elliott C, Newman N, Madon A. Gentamicin effects on Kidney Dis. 1990;15:583-591.
cotherapy. 1995;15:625-630. urinary electrolyte excretion in healthy subjects. Clin Phar- 299. Rodriquez HJ, Hogan WC, Hellman RN, et al. Mechanism
244. Dowling TC, Wang ES, Ferrucci L, et al. Glomerular filtra- macol Ther. 2000;67:16-21. of activation of renal Na+-K+-ATPase in the rat: effects of
tion rate equations overestimate creatinine clearance in 272. Bennett WM, Elliott WC, Houghton DC, et al. Reduction potassium loading. Am J Physiol. 1980;238:F315-F323.
older individuals enrolled in the Baltimore longitudinal of experimental gentamicin nephrotoxicity in rats by 300. Cronin RE, Newman JA. Protective effect of thyroxine but
study on aging: impact on renal drug dosing. Pharmaco- dietary calcium loading. Antimicrob Agents Chemother. not parathyroidectomy on gentamicin nephrotoxicity. Am
therapy. 2013;33:912-921. 1982;22:508-512. J Physiol. 1985;248:F332-F339.
245. Gilbert DN, Bennett WM. Use of antimicrobial agents in 273. Quarum ML, Houghton DC, Gilbert DN, et al. Increasing 301. Adelman RD, Spangler WL, Beasom F, et al. Furosemide
renal failure. Infect Dis Clin North Am. 1989;3:517-531. dietary calcium moderates experimental gentamicin neph- enhancement of experimental gentamicin nephrotoxicity:
246. Chuck SK, Raber SR, Rodvold KA, et al. National survey rotoxicity. J Lab Clin Med. 1984;103:104-114. comparison of functional and morphological changes with
of extended-interval aminoglycoside dosing. Clin Infect 274. Humes HD, Sastrasink M, Weinberg JM. Calcium is a com- activities of urinary enzymes. J Infect Dis. 1979;140:
Dis. 2000;30:433-439. petitive inhibitor of gentamicin renal membrane binding 342-352.
247. Aronoff GR, ed. Drug Prescribing in Renal Failure. 4th ed. interactions and dietary calcium supplementation protects 302. Vakil N, Abu-Alfa A, Mujais SK. Gentamicin nephrotoxic-
Philadelphia: American College of Physicians; 1999. against gentamicin nephrotoxicity. J Clin Invest. 1984;73: ity in extrahepatic cholestasis: modulation by dietary
248. Reetze-Bonorden P, Bohler J, Keller E. Drug dosage in 134-147. calcium. Hepatology. 1989;9:519-524.
patients during continuous renal replacement therapy. Clin 275. Ernst S. Model of gentamicin induced nephrotoxicity and 303. Camps J, Sola X, Rimola A, et al. Comparative study of
Pharmacokinet. 1993;24:362-379. its amelioration by calcium and thyroxine. Med Hypoth. aminoglycoside nephrotoxicity in normal rats and rats with
249. McCormack JP, Jewesson PJ. A critical reevaluation of the 1989;30:195-202. experimental cirrhosis. Hepatology. 1988;8:837-844.
“therapeutic range” of aminoglycoside. Clin Infect Dis. 276. Mizuta K, Saito A, Watanabe T, et al. Ultrastructural local- 304. Lietman PS. Liver disease, aminoglycoside antibiotics, and
1992;14:320-329. ization of megalin in the rat cochlear duct. Hear Res. 1999; renal dysfunction. Hepatology. 1988;4:966-968.
250. Edwards C, Bent AJ, Venables CW, et al. Sampling time for 129:83-91. 305. Teixeira RB, Kelley J, Alpert H, et al. Complete protection
serum gentamicin levels. J Antimicrob Chemother. 277. Scherbeich JE, Mondorf WA. Nephrotoxic potential of from gentamicin-induced acute renal failure in the diabetes
1992;29:575-578. antiinfective drugs as assessed by tissue-specific protein- mellitus rat. Kidney Int. 1982;21:600-612.
251. Dager WE, King JH. Aminoglycosides in intermittent uria of renal antigens. Int J Clin Pharm Ther. 1998;36: 306. Elliott WB, Houghton DC, Gilbert DN, et al. Experimental
hemodialysis: pharmacokinetics with individual dosing. 152-158. gentamicin nephrotoxicity: effect of streptozotocin-
Ann Pharmacother. 2006;20:9-14. 278. Lipsky JJ, Lietman PS. Neomycin inhibition of adenosine induced diabetes. J Pharmacol Exp Ther. 1985;233:
252. Pai MP, Nafziger AN, Bertino JS. Simplified estimation of triphosphate: evidence for a neomycin phospholipid 264-270.
aminoglycoside pharmacokinetics in underweight and interaction. Antimicrob Agents Chemother. 1980;18: 307. Wood CA, Kohlhepp SJ, Kohnen PW, et al. Vancomycin
obese adult patients. Antimicrob Agents Chemother. 2011; 532-535. enhancement of experimental tobramycin nephrotoxicity.
55:4006-4011. 279. Sorribas V, Halaikel N, Puttaparthi K, et al. Gentamicin Antimicrob Agents Chemother. 1985;30:20-24.
253. Traynor AM, Nafziger AN, Bertino JS. Aminoglycoside causes endocytosis of Na/Pi cotransporter protein (NaPi- 308. Kohlhepp SJ, Gilbert DN, Kohnen PW, et al. Teicoplanin
dosing weight correction factors for patients of various 2). Kidney Int. 2001;59:1024-1036. enhancement of experimental tobramycin nephrotoxicity
body sizes. Antimicrob Agents Chemother. 1995;39: 280. von Vigier RO, Truttman AC, Zindler-Schmocker K, et al. [abstract]. Proceedings of the 31st Interscience Conference on
545-548. Aminoglycosides and renal magnesium homeostasis in Antimicrobial Agents Chemotherapy. Washington, DC:
254. Levey AS, Bosch JP, Lewis JB, et al. A more accurate humans. Nephrol Dial Transplant. 2000;15:822-826. American Society for Microbiology; 1991.
method to estimate glomerular filtration rate from serum 281. Mohammed EM, Laurent G, Mingeot-Leclercq M-P, et al. 309. Golper TA, Noonan HM, Elzinga L, et al. Vancomycin
creatinine: new prediction equation. Ann Intern Med. Apoptosis in renal proximal tubules of rats treated with low pharmacokinetics, renal handling and non-renal clearance
1999;130:461-470. does of aminoglycosides. Antimicrob Agents Chemother. in normal human subjects. Clin Pharmacol Ther. 1988;43:
255. Charhon N, Neely MN, Bourguignon L, et al. Comparison 2000;44:665-675. 565-570.
of four renal function estimation equations for pharmaco- 282. Appel GB. Aminoglycoside nephrotoxicity. Am J Med. 310. English J, Gilbert DN, Kohlhepp SJ, et al. Attenuation of
kinetic modeling of gentamicin in geriatric patients. Anti- 1990;88(suppl C):S16-S20. experimental tobramycin nephrotoxicity by ticarcillin.
microb Agents Chemother. 2012;56:1862-1869. 283. Gilbert DN, Houghton DC, Bennett WM, et al. Revers- Antimicrob Agents Chemother. 1985;276:897-902.
256. Wallace AW, Jones M, Bertino JS. Evaluation of four once- ibility of gentamicin nephrotoxicity in rats: recovery during 311. Sabra R, Branch RA. Role of sodium in protection by
daily aminoglycoside dosing nomograms. Pharmacother- continuous drug administration. Proc Soc Exp Biol Med. extended spectrum penicillins against tobramycin-induced
apy. 2002;22:1077-1083. 1979;160:99-103. nephrotoxicity. Antimicrob Agents Chemother. 1990;340:
257. MacDonald I, Staatz CE, Jelliffe RW, et al. Evaluation and 284. Luft FC, Bennett WM, Gilbert DN. Experimental amino- 1020-1025.
comparison of simple multiple model richer data multiple glycoside nephrotoxicity: accomplishments and future 312. Barr GA, Mazze RI, Cousins MJ, et al. An animal model
model, and sequential interacting multiple model (IMM) potential. Rev Infect Dis. 1983;5(suppl 2):S268-S293. for combined methoxyflurane and gentamicin nephrotox-
Bayesian analysis of gentamicin and vancomycin data col- 285. Gilbert DN, Plamp C, Starr P, et al. Comparative nephro- icity. Br J Anaesth. 1973;45:306-331.
lected from patients undergoing cardiothoracic surgery. toxicity of gentamicin and tobramycin in rats. Antimicrob 313. Whiting PH, Simpson JG. The enhancement of
Ther Drug Monit. 2008;30:67-74. Agents Chemother. 1979;13:34-40. cyclosporin-A induced nephrotoxicity by gentamicin.
258. Streetman DS, Nafziger AN, Destache CJ, et al. Individual- 286. Sandoval RM, Reilly JP, Running W, et al. A non- Biochem Pharmacol. 1983;32:2025-2028.
ized pharmacokinetic monitoring results in less nephrotoxic gentamicin congener that retains antimicro- 314. Ryffel B, Muller AM, Mihatsch MJ. Experimental cyclospo-
aminoglycoside-associated nephrotoxicity and fewer asso- bial efficacy. J Am Soc Nephrol. 2006;17:2697-2705. rine nephrotoxicity: risk of concomitant chemotherapy.
ciated costs. Pharmacotherapy. 2001;21:443-451. 287. Denamu S, Van Bambeke F, Mingeot-Leclercq MP, et al. Clin Nephrol. 1988;25(suppl 1):S121-S125.
259. Reinhard MK, Hottendorf GH, Powell FD. Differences in Apoptosis induced by aminoglycosides in LCC-PK1 cells: 315. Jongejan HTM, Provoost AP, Molenaar JC. Potentiated
the sensitivity of Fischer and Sprague-Dawley rats to ami- comparative study of neomycin, gentamicin, amikacin, and nephrotoxicity of cisplatin when combined with amikacin
noglycoside nephrotoxicity. Toxicol Pathol. 1991;19:66-71. isepamicin using electroporation. Antimicrob Agents Che- comparing young and adult rats. Pediatr Nephrol. 1989;
260. Mingeot-Leclercq M-P, Tulkens PM. Aminoglycosides: mother. 2008;52:2236-2238. 31:290-295.
nephrotoxicity. Antimicrob Agents Chemother. 1999;43: 288. Bennett WM, Plamp CE, Gilbert DN, et al. The influence 316. Williams PD, Hottdenford GH. Inhibition of renal mem-
1005-1012. of dosage regimen on experimental gentamicin nephrotox- brane binding and nephrotoxicity of gentamicin by polyas-
261. Verroust PJ, Birn H, Nielsen R, et al. The tandem endocytic icity: dissociation of peak serum levels from renal failure. paragine and polyaspartic acid in the rat. Res Commun
receptors megalin and cubilin are important proteins in J Infect Dis. 1979;140:576-580. Chem Pathol Pharmacol. 1985;47:317-320.
renal pathology. Kidney Int. 2002;62:745-756. 289. Powell SH, Thompson WL, Luthe MA, et al. Once daily vs 317. Gilbert DN, Wood CA, Kohlhepp SJ, et al. Polyaspartic
262. Sandoval RM, Molitoris BA. Gentamicin traffics retrograde continuous aminoglycoside dosing: efficacy and toxicity in acid prevents experimental aminoglycoside nephrotoxicity.
through the secretory pathway and is released in the animal and clinical studies of gentamicin, netilmicin, and J Infect Dis. 1989;159:945-953.
cytosol via the endoplasmic reticulum. Am J Physiol Renal tobramycin. J Infect Dis. 1983;147:918-923. 318. Swan SK, Kohlhepp SJ, Kohnen PW, et al. Long-term pro-
Physiol. 2004;286:F617-F624. 290. Beauchamp D, Gourde P, Thereault G, et al. Age-dependent tection of polyaspartic acid in experimental gentamicin
263. Schmitz C, Hilpert J, Jacobsen C, et al. Megalin deficiency gentamicin experimental nephrotoxicity. J Pharmacol Exp nephrotoxicity. Antimicrob Agents Chemother. 1991;35:
offers protection from renal aminoglycoside accumulation. Ther. 1992;260:444-449. 2591-2595.
J Biol Chem. 2002;277:618-622. 291. Bennett WM, Parker RA, Elliott WB, et al. Sex: a determi- 319. Gilbert DN, Kohlhepp SJ, Swan SK, et al. Pharmacologic
264. Verroust PJ, Christensen EI. Megalin and cubilin: the story nant of susceptibility to gentamicin nephrotoxicity in the limits of polyaspartic acid’s protective effect on experimen-
of two multipurpose receptors unfolds. Nephrol Dial Trans- rat. J Infect Dis. 1982;145:370-373. tal gentamicin nephrotoxicity. Antimicrob Agents Che-
plant. 2002;17:1867-1871. 292. Moore RD, Smith CR, Lipsky JJ, et al. Risk factors for mother. 1993;37:347-348.
265. Sandoval RM, Dunn KW, Molitoris BA. Gentamicin traffics nephrotoxicity in patients treated with aminoglycosides. 320. Swan SK, Gilbert DN, Kohlhepp SJ, et al. Duration of the
rapidly and directly to the Golgi complex in LLC-PK1 cells. Ann Intern Med. 1984;100:352-357. protective effect of polyaspartic acid on experimental gen-
Am J Physiol. 2000;279:F884-F890. 293. Sawyers CL, Moore RD, Lerner SA, et al. A model for tamicin nephrotoxicity. Antimicrob Agents Chemother.
266. Bennett WM, Mela-Riker L, Houghton DC, et al. Micro- predicting nephrotoxicity in patients treated with amino- 1992;36:2556-2558.
somal protein synthesis inhibition: an early manifestation glycosides. J Infect Dis. 1985;153:1062-1068. 321. Ramsammy LS, Josepovitz C, Lane BP, et al. Polyaspartic
of gentamicin nephrotoxicity. Am J Physiol. 1988;255: 294. Elliott WB, Parker RA, Houghton DC, et al. Effect of acid protects against gentamicin nephrotoxicity in the rat.
F265-F269. sodium bicarbonate and ammonium chloride ingestion in J Pharmacol Exp Ther. 1989;250:149-153.
267. Gilbert DN, Bennett WM. Progress in the elucidation of experimental gentamicin nephrotoxicity in rats. Res 322. Beauchamp D, Laurent G, Maldague P, et al. Protection
aminoglycoside nephrotoxicity. Contemp Issues Infect Dis. Commun Pathol Pharmacol. 1980;28:483-496. against gentamicin-induced early renal alterations (phos-
1984;1:121-152. 295. Bennett WM, Hartnett MN, Gilbert D, et al. Effect of pholipidosis and increased DNA synthesis) by coadminis-
268. Buss WC, Piatt MK. Gentamicin administered in vivo sodium intake on gentamicin nephrotoxicity in the rat. tration of poly-l-aspartic acid. J Pharmacol Exp Ther.
reduces protein synthesis in microsomes subsequently Proc Soc Exp Biol Med. 1976;151:736-738. 1990;255:858-866.
321.e5
323. Kishore BK, Ibrahim S, Lambrict P, et al. Comparative 350. Klastersky J, Hensgens C, Debusscher I. Empiric therapy 380. Vandebona H, Mitchell P, Manwaring N, et al. Prevalence
assessment of poly-L-aspartic and poly-L-glutamic acids as for cancer patients: comparative study of ticarcillin- of mitochondrial 1555 A-to-G mutation in adults of Euro-
protectants against gentamicin-induced renal lysosomal tobramycin, ticarcillin-cephalothin, and cephalothin- pean descent. N Engl J Med. 2009;360:642-644.
phospholipidosis, phospholipiduria and cell proliferation tobramycin. Antimicrob Agents Chemother. 1975;7: 381. Chaig MR, Zernotti ME, Soria NW, et al. A mutation in

Chapter 25  Aminoglycosides


in rats. J Pharmacol Exp Ther. 1992;262:424-432. 640-645. mitochondrial 12S rRNA, A827G, in Argentinean family
324. Kohlhepp SJ, McGregor D, Gilbert DN. Determinants of 351. Wade JC, Smith CR, Petty BG, et al. Cephalothin plus an with hearing loss after aminoglycoside treatment. Biochem
the in vitro interaction of polyaspartic acid and aminogly- aminoglycoside is more nephrotoxic than methicillin plus Biophys Res Commun. 2008;368:631-636.
coside antibiotics. J Pharmacol Exp Ther. 1992;263: an aminoglycoside. Lancet. 1978;3:604-606. 382. Tang H-Y, Hucheson E, Neill S, et al. Genetic susceptibility
1464-1470. 352. Nix DE, Thomas JK, Symonds WT, et al. Assessment of the to aminoglycoside ototoxicity: how many are at risk? Genet
325. Ramsammy L, Josepovitz C, Lane B, et al. Polyaspartic acid enzymuria resulting from gentamicin alone and combina- Med. 2002;4:336-345.
inhibits gentamicin-induced perturbations of phospho- tions of gentamicin with various β-lactam antibiotics. Ann 383. Zhu Y, Qian Y, Tang X, et al. Aminoglycoside-induced and
lipid metabolism. Am J Physiol. 1990;58:C1141-C1149. Pharmacother. 1997;31:696-703. non-syndromic hearing loss is associated with the G7444A
326. Luft FC. Cephalosporin and aminoglycoside interactions: 353. Farber BF, Moellering RC. Retrospective study of the toxic- mutation in the mitochondrial COI/tRNASer (UCN) genes
clinical and toxicologic implications. In: Whelton A, Neu ity of preparations of vancomycin from 1974-1981. Antimi- in two Chinese families. Biochem Biophys Res Commun.
HC, eds. The Aminoglycosides. New York: Marcel Dekker; crob Agents Chemother. 1983;23:138-141. 2006;342:843-850.
1982:387-399. 354. Rybak MJ, Albrecht LM, Boike SC, et al. Nephrotoxicity of 384. Wang Q, Li QZ, Han D, et al. Clinical and molecular
327. Yanagida C, Ito K, Koniya I, et al. Protective effect of fos- vancomycin alone and with an aminoglycoside. J Antimi- analysis of a four-generation Chinese family with
fomycin on gentamicin-induced lipid peroxidation of rat crob Chemother. 1990;25:679-687. aminoglycoside-induced and nonsyndromic hearing loss
renal tissue. Chem Biol Interact. 2004;148:139-147. 355. Paul M, Schlesinger A, Grozinsky-Glasberg S, et al. associated with the mitochondrial 12S rRNA C1494T
328. Bertino JS Jr, Booker LA, Franck PA, et al. Incidence of β-lactam versus β-lactam-aminoglycoside combination mutation. Biochem Biophys Res Commun. 2006;340:
and significant risk factors for aminoglycoside-associated therapy in cancer patients with neutropenia. Cochrane 583-588.
nephrotoxicity in patients dosed by using individualized Database Syst Rev. 2010;(3):CD003038. 385. Hobbie SN, Bruell CM, Akshay S, et al. Mitochondrial
pharmacokinetic monitoring. J Infect Dis. 1993;167: 356. Xie J, Talaska AE, Schacht J. New developments in amino- deafness alleles confer misreading of the genetic code. Proc
173-179. glycoside therapy and ototoxicity. Hear Res. 2011;281:28-37. Natl Acad Sci U S A. 2008;105:3244-3249.
329. Kahlmeter G, Dahlager JI. Aminoglycoside toxicity: a 357. Hinshaw HC, Feldman WH. Streptomycin in treatment of 386. Veenstra DL, Harris J, Gibson RL, et al. Pharmacogenomic
review of clinical studies published between 1975 and 1982. clinical tuberculosis: a preliminary report. Proc Staff Meet testing to prevent aminoglycoside-induced hearing loss
J Antimicrob Chemother. 1984;13(suppl A):S9-S22. Mayo Clin. 1945;20:313-318. in cystic fibrosis patients: potential impact on clinical,
330. Lietman PS, Smith CR. Aminoglycoside nephrotoxicity in 358. Jackson GG, Finland M. International Symposium on Gen- patient, and economic outcomes. Genet Med. 2007;9:
humans. Rev Infect Dis. 1983;5(suppl 2):S284-S292. tamicin. J Infect Dis. 1969;119:341-540. 695-704.
331. Smith CR, Baughman KL, Edwards CQ, et al. Controlled 359. Finland M, Hewitt WL. Second International Symposium 387. Hamasaki K, Rando RR. Specific binding of aminoglyco-
comparison of amikacin and gentamicin. N Engl J Med. on Gentamicin. J Infect Dis. 1971;124:S1-S300. side to a human rRNA construct based on a DNA poly-
1977;296:349-353. 360. Brummett RE, Fox KE. Aminoglycoside induced hearing morphism which causes aminoglycoside-induced deafness.
332. Smith CR, Lipsky JJ, Laskin OL, et al. Double-blind com- loss in humans. Antimicrob Agents Chemother. 1989;33: Biochemistry. 1997;36:12323-12328.
parison of the nephrotoxicity and auditory toxicity of gen- 797-800. 388. Xing G, Chen Z, Wei Q, et al. Mitochondrial 12S rRNA
tamicin and tobramycin. N Engl J Med. 1980;302: 361. Ahmed RM, Hannigan IP, MacDougall HG, et al. Genta- A827G mutation is involved in the genetic susceptibility to
1106-1109. micin ototoxicity: a 23-year selected case series of 103 aminoglycoside ototoxicity. Biochem Biophys Res Commun.
333. Smith CR, Ambinder R, Lipsky JJ, et al. Cefotaxime com- patients. Med J Aust. 2012;196:701-704. 2006;346:1131-1135.
pared with nafcillin plus tobramycin for serious bacterial 362. Fausti SA, Henry JA, Schaffer HI, et al. High-frequency 389. Beaubien AR, Ormsby E, Bayne A, et al. Evidence that
infections. Ann Intern Med. 1984;101:469-477. audiometric monitoring for early detection of aminoglyco- amikacin ototoxicity is related to total perilymph area
334. Bertino JS, Booker LA, Franck PA, et al. Incidence of and side ototoxicity. J Infect Dis. 1992;165:1026-1032. under the concentration-time curve regardless of concen-
significant risk factors for aminoglycoside-associated neph- 363. Govaerts PJ, Claes PH, DeHeyring PHV, et al. tration. Antimicrob Agents Chemother. 1991;35:1070-1074.
rotoxicity in patients dosed by using individualized phar- Aminoglycoside-induced ototoxicity. Toxicol Lett. 1990;52: 390. Dille MF, Konrad-Martin D, Gallun F, et al. Tinnitus onset
macokinetic monitoring. J Infect Dis. 1993;167:173-179. 227-251. rates from chemotherapeutic agents and ototoxic antibiot-
335. Lerner AM, Cone LA, Jansen W, et al. Randomized, con- 364. Dulon D, Aran JM, Zajic G, et al. Comparative uptake of ics: results of a large prospective study. J Am Acad Audiol.
trolled trial of the comparative efficacy, auditory toxicity gentamicin, netilmicin, and amikacin in the guinea pig 2010;21:409-417.
and nephrotoxicity of tobramycin and netilmicin. Lancet. cochlea and vestibule. Antimicrob Agents Chemother. 1986; 391. Sataloff J, Wagner S, Menduke H. Kanamycin ototoxicity in
1983;1:1123-1126. 30:96-100. healthy men. Arch Otolaryngol. 1964;80:413-417.
336. Lerner SA, Schmitt BA, Seligsohn R, et al. Comparative 365. Marcotti W, vanNetten SM, Kros CJ. The aminoglycoside 392. Davey PG, Jabeen F, Harpur ES, et al. The use of pure-tone
study of ototoxicity and nephrotoxicity in patients ran- antibiotic dihydrostreptomycin rapidly enters hair cells audiometry in the assessment of gentamicin auditory tox-
domly assigned treatment with amikacin and gentamicin. through the mechano-declinical transducer channel. icity. Br J Audiol. 1982;16:151-154.
Am J Med. 1986;80(suppl 5B):S98-S104. J Physiol. 2005;567:505-521. 393. Moore RD, Smith CR, Lietman PS. Risk factors for the
337. Hou SH, Bushinsky DA, Wish JB, et al. Hospital-acquired 366. de Groot JC, Meeuwsen F, Ruizendaal WE, et al. Ultra- development of auditory toxicity in patients receiving ami-
renal insufficiency: a prospective study. Am J Med. 1983; structural localization of gentamicin in the cochlear. Hear noglycosides. J Infect Dis. 1984;149:23-30.
74:243-248. Res. 1990;50:35-42. 394. Brummett RE, Fox KE. Vancomycin- and erythromycin-
338. Trollfors B. Gentamicin-associated changes in renal func- 367. Hashino E, Shero M, Salvi RJ. Lysosomal targeting and induced hearing loss in humans. Antimicrob Agents Che-
tion reversible during continued treatment. J Antimicrob accumulation of aminoglycoside antibiotics in sensory hair mother. 1989;33:791-796.
Chemother. 1983;12:285-287. cells. Brain Res. 1997;777:75-85. 395. Tan C-T, Hsu C-J, Lee S-Y, et al. Potentiation of noise-
339. Gilbert DN. Meta-analyses are no longer required for 368. Wang Q, Steyger PS. Trafficking of systemic fluorescent induced hearing loss by amikacin in guinea pigs. Hear Res.
determining the efficacy of single daily dosing of amino- gentamicin into the cochlea and hair cells. J Assoc Res Oto- 2001;161:72-80.
glycosides. Clin Infect Dis. 1997;24:816-819. laryngol. 2009;10:205-219. 396. Brummett RE, Fox KE, Kempton JB. Quantitative relation-
340. Buchholtz K, Larsen CT, Hassager C, et al. Severity of gen- 369. Hutchin T, Cortopassi G. Proposed molecular and cellular ships of the interaction between sound and kanamycin.
tamicin’s nephrotoxic effect on patients with infective mechanism of aminoglycoside ototoxicity. Antimicrob Arch Otolaryngol Head Neck Surg. 1992;118:498-500.
endocarditis: a prospective observational cohort study of Agents Chemother. 1994;38:2517-2520. 397. Tran Ba Huy PT, Deffrennes D. Aminoglycoside ototoxic-
373 patients. Clin Infect Dis. 2009;48:65-71. 370. Tran Ba Huy P, Bernard P, Schacht J. Kinetics of gentamicin ity: influence of dosage regimen on drug uptake and cor-
341. Lepper PM, Held TK, Schneider EM, et al. Clinical implica- uptake and release in the rat: comparison of inner ear relation between membrane binding and some clinical
tions of antibiotic-induced endotoxin release in septic tissues and fluids with other organs. J Clin Invest. 1986; features. Acta Otolaryngol. 1988;105:511-515.
shock. Intensive Care Med. 2002;28:824-833. 77:1492-1500. 398. Moore RD, Smith CR, Lietman PS. Risk factors for the
342. Sjolin J, Goscinski G, Lundhom M, et al. Endotoxin release 371. Guthrie OW. Aminoglycoside induced toxicity. Toxicology. development of auditory toxicity in patients receiving ami-
from Escherichia coli after exposure to tobramycin: dose- 2008;249:91-96. noglycosides. J Infect Dis. 1984;149:23-30.
dependency and reduction in cefuroxime-induced endo- 372. Karasawa T, Steyger PS. Intracellular mechanisms of 399. Gatell JM, Ferran F, Araujo V, et al. Univariate and multi-
toxin release. Clin Microbiol Infect. 2000;6:74-81. aminoglycoside-induced cytotoxicity. Integr Biol. 2011;3: variate analyses of risk factors predisposing to auditory
343. Green FJ, Lavelle KJ, Aronoff GR, et al. Management of 879-886. toxicity in patients receiving aminoglycosides. Antimicrob
amikacin overdose. Am J Kidney Dis. 1981;1:110-112. 373. Forge A, Schacht J. Aminoglycoside antibiotics. Audiol Agents Chemother. 1987;31:1383-1387.
344. Ho PW, Pien FD, Kominami N. Massive amikacin “over- Neurootol. 2000;5:3-22. 400. Buring JE, Evans DA, Mayrent SL, et al. Randomized trials
dose.” Ann Intern Med. 1979;91:227-228. 374. Takumida M, Anniko M, Shimizu A, et al. Neuroprotection of aminoglycoside antibiotics: quantitative overview. Rev
345. Nahata MC. Lack of nephrotoxicity in pediatric patients of vestibular sensory cells from gentamicin ototoxicity Infect Dis. 1988;10:951-957.
receiving vancomycin and aminoglycoside therapy. Che- obtained using nitric oxide synthase inhibitors, reactive 401. Moore RD, Lerner SA, Levine DP. Nephrotoxicity and oto-
motherapy. 1987;33:302-304. oxygen species scavengers, brain-derived neurotrophic toxicity of aztreonam versus aminoglycoside therapy in
346. Cooper B, Creger RJ, Soegiarso W, et al. Renal dysfunction factors and calpain inhibitors. Acta Otolaryngol. 2003;123: seriously ill non-neutropenic patients. J Infect Dis. 1992;
during high-dose cisplatin therapy and autologous hema- 8-13. 165:683-688.
topoietic stem cell transplantation: effect of aminoglyco- 375. Hashino E, Tanaka Y, Salvi RJ, et al. Hair cell regeneration 402. Manian FA, Stone WJ, Alford R. Adverse antibiotic effects
side therapy. Am J Med. 1993;94:497-504. in the adult budgerigar after kanamycin ototoxicity. Hear associated with renal insufficiency. Rev Infect Dis. 1990;12:
347. Leach CT, Kuhls TL, Brill JE, et al. Use of aminoglycosides Res. 1992;59:46-58. 236-249.
during cyclosporine A immunosuppression after liver 376. Lombarte A, Yan HY, Popper AN, et al. Damage and regen- 403. Sha S-H, Qiu JH, Schacht J. Aspirin attenuates gentamicin-
transplantation in children. Pediatr Infect Dis J. 1989;8: eration of hair cell ciliary bundles in a fish ear following induced hearing loss. N Engl J Med. 2006;354:1856-1857.
354-357. treatment with gentamicin. Hear Res. 1993;64:166-174. 404. Amiko M, Bagger-Sjoback D, Wersall J, et al. Gentamicin
348. Mazze RI, Cousins MJ. Combined nephrotoxicity of genta- 377. Forge A, Li L, Corwin JT, et al. Ultrastructural evidence for binding to the isolated crista ampullaris of the guinea pig.
micin and methoxyflurane in man. Br J Anaesth. 1973;45: hair cell regeneration in the mammalian inner ear. Science. Res Commun Chem Pathol Pharmacol. 1982;37:333-342.
394-398. 1993;259:1616-1621. 405. Minor LB. Gentamicin-induced bilateral vestibular hypo-
349. Schimpff SC, Gaya H, Klastersky J, et al. Three antibiotic 378. Fischel-Ghodsian N. Genetic factors in aminoglycoside function. JAMA. 1998;279:541-544.
regimens in the treatment of infection in febrile granulo- toxicity. Ann N Y Acad Sci. 1999;884:99-109. 406. Black FO, Gianna-Poulin C, Pesznecker SC. Recovery from
cytopenic patients with cancer. The EORTC International 379. Bitner-Glindzicz M, Pembrey M, Duncan A, et al. Preva- vestibular ototoxicity. Otol Neurol. 2001;22:662-671.
Antimicrobial Therapy Project Group. J Infect Dis. 1978; lence of mitochondrial 1555 A-to-G mutation in European 407. Fee WE. Aminoglycoside ototoxicity in the human. Laryn-
137:14-29. children. N Engl J Med. 2009;360:640-642. goscope. 1980;90(suppl 24):1-9.
321.e6
408. Walsh RM, Bath AP, Bance ML. Reversible tobramycin- 439. Hewitt WL, Young LS. Symposium perspective. Am J Med. 464. Ehrmann S, Mercier E, Vecellio L, et al. Pharmacokinetics
induced bilateral high-frequency vestibular toxicity. ORL J 1977;62:863-867. of high-dose nebulized amikacin in mechanically venti-
Otorhinolaryngol Relat Spec. 2000;62:156-159. 440. Freundlich M, Thomsen RW, Pedersen L. Aminoglycoside lated healthy subjects. Intensive Care Med. 2008;34:
409. Forge A, Li L, Corwin JT, et al. Ultrastructural evidence for treatment and mortality after bacteraemia in patients given 755-762.
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

hair-cell regeneration in the mammalian inner ear. Science. appropriate empirical therapy: a Danish hospital-based 465. Falagas ME, Matthaiou DK, Karveli EA, et al. Meta-
1993;259:1616-1619. cohort study. J Antimicrob Chemother. 2007;60:1115-1123. analysis: randomized controlled trials of clindamicin/
410. Rubel EW, Dew LA, Roberson DW. Mammalian vestibular 441. Bliziotis IA, Michalopoulos A, Kasiakou SK, et al. Cipro- aminoglycoside vs β-lactam monotherapy for the treat-
hair-cell regeneration. Science. 1995;267:701-703. floxacin vs aminoglycoside in combination with a beta- ment of intra-abdominal infections. Aliment Pharmacol
411. Harner SG, Driscoll CL, Facer GW, et al. Long-term fol- lactam for the treatment of febrile neutropenia: a Ther. 2006;25:537-556.
lowup of transtympanic gentamicin for Meniere’s syn- meta-analysis of randomized controlled trials. Mayo Clin 466. van Nieuwkoop C, den Exter PL, Elzevier HW, et al. Intra-
drome. Otol Neurol. 2001;22:210-214. Proc. 2005;80:1122-1125. vesical gentamicin for recurrent urinary tract infection in
412. Pullens B, van Benthem PP. Intratympanic gentamicin for 442. Leibovici L, Paul M, Poznanski O, et al. Monotherapy patients with intermittent bladder catheterization. Int J
Meniere’s disease or syndrome. Cochrane Database Syst versus β-lactam-aminoglycoside combination treatment Antimicrob Agents. 2010;36:485-490.
Rev. 2011;(3):CD008234. for gram-negative bacteria: a prospective, observational 467. Zebner R, Quinn JP. Antimicrobials in cystic fibrosis:
413. Perez N, Martin E, Garcia-Tapia R. Intratympanic genta- study. Antimicrob Agents Chemother. 1997;41:1127-1133. emergence of resistance and implications for treatment.
micin for intractable Meniere’s disease. Laryngoscope. 2003; 443. Marcus R, Paul M, Elphick H, et al. Clinical implications Semin Respir Infect. 1992;7:210-217.
113:456-464. of β-lactam-aminoglycoside synergism: systematic review 468. deGroot R, Smith AL. Antibiotic pharmacokinetics in
414. Snavely SR, Hodges GR. The neurotoxicity of antibacterial of randomised trials. Internat J Antimicrobial Agents. 2011; cystic fibrosis: differences and clinical significance. Clin
agents. Ann Intern Med. 1984;101:92-104. 37:491-503. Pharmacokinet. 1987;13:228-253.
415. Talbot PA. Potentiation of aminoglycoside-induced neuro- 444. Tamma PD, Cosgrove SE, Maragakis LL. Combination 469. Tan K, Bunn H. Once daily versus multiple daily dosing
muscular blockade by protons in vitro and in vivo. J Phar- therapy for treatment of infections with gram-negative with intravenous aminoglycosides for cystic fibrosis.
macol Exp Ther. 1987;241:686-694. bacteria. Clin Microbiol Rev. 2012;25:450-470. Cochrane Database Syst Rev. 2000;(4):CD002009.
416. Del-Pozo E, Baezem JM. Effects of calcium channel block- 445. Martinez JA, Cobos-Triqueros N, Soriano A, et al. Influ- 470. Tan KH, Mulheran M, Knox AJ, Smyth AR. Aminoglyco-
ers on neuromuscular blockade induced by aminoglyco- ence of empiric therapy with a beta-lactam alone or com- side prescribing and surveillance in cystic fibrosis. Am J
side antibiotics. Eur J Pharmacol. 1986;128:49-54. bined with an aminoglycoside on prognosis of bacteremia Respir Crit Care Med. 2003;167:819-823.
417. Gay CT, Marks WA, Riley HD Jr, et al. Infantile botulism. due to gram-negative microorganisms. Antimicrob Agents 471. Mulheran M, Degg C, Morgan DW, et al. The occurrence
South Med J. 1988;81:457-460. Chemother. 2010;54:3590-3596. and risk of cochleotoxicity in cystic fibrosis patients receiv-
418. Hokkanen E. The aggravating effect of some antibiotics on 446. Micek ST, Welch EC, Kha J, et al. Empiric combination ing repeated high dose aminoglycoside therapy. Antimicrob
the neuromuscular blockade in myasthenia gravis. Acta antibiotic therapy is associated with improved outcome Agents Chemother. 2001;45:2502-2509.
Neurol Scand. 1964;40:346-352. against sepsis due to gram-negative bacteria: a retro­ 472. Elphick HE, Tan A. Single versus combination intravenous
419. Warner WA, Sanders E. Neuromuscular blockade asso­ spective analysis. Antimicrob Agents Chemother. 2010;54: antibiotic therapy for people with cystic fibrosis. Cochrane
ciated with gentamicin therapy. JAMA. 1971;215: 1742-1748. Database Syst Rev. 2005;(2):CD002007.
1153-1154. 447. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis 473. Ryan G, Mukhopadhyay S, Singh M. Nebulized anti-
420. Holtzman JL. Gentamicin and neuromuscular blockade. campaign: international guidelines for management of pseudomonal antibiotics for cystic fibrosis. Cochrane Data-
Ann Intern Med. 1976;84:55. severe sepsis and septic shock: 2012. Crit Care Med. 2013; base Syst Rev. 2003;(3):CD001021.
421. Wong J, Brown G. Does once-daily dosing of aminoglyco- 41:580-637. 474. Smyth AR, Bhatt J, Tan KH. Once-daily versus multiply-
sides affect neuromuscular function? J Clin Pharm Ther. 448. Graham JC, Gould FK. Role of aminoglycosides in the daily dosing with intravenous aminoglycosides for cystic
1996;21:407-411. treatment of bacterial endocarditis. J Antimicrob Che- fibrosis. Cochrane Database Syst Rev. 2006;(3):CD002009.
422. Weber-Carstens S, Deja M, Koch S, et al. Rick factors in mother. 2002;49:437-444. 475. Breen L, Aswani N. Elective versus symptomatic intrave-
critical illness myopathy during the early course of critical 449. Falagas ME, Matthaiou DK, Bliziotis IA. The role of ami- nous antibiotic therapy for cystic fibrosis. Cochrane Data-
illness: a prospective observational study. Crit Care. 2010; noglycosides in combination with a beta-lactam for the base Syst Rev. 2001;(4):CD002767.
14:R119. treatment of bacterial endocarditis: a meta-analysis of com- 476. Mouton JW, Jacobs N, Tiddens H, et al. Pharmacodynam-
423. Wright JM, Collier B. The effects of neomycin upon trans- parative trials. J Antimicrob Chemother. 2006;57:639-647. ics of tobramycin in patients with cystic fibrosis. Diagn
mitter release and action. J Pharmacol Exp Ther. 1977;200: 450. Buchholtz K, Larsen CT, Schaadt B, et al. Once versus twice Microbiol Infect Dis. 2005;52:123-127.
576-587. daily gentamicin dosing for infective endocarditis: a ran- 477. Burkhardt O, Lehmann C, Madabush R, et al. Once-daily
424. Lee C, DeSilva AJ. Acute and subchronic neuromuscular domized clinical trial. Cardiology. 2011;119:65-71. tobramycin in cystic fibrosis: better for clinical outcomes
blocking characteristics of streptomycin: a comparison 451. Baldous LM, Wilson WR, Bayer AS, et al. Infective endo- than thrice-daily tobramycin but more resistance develop-
with neomycin. Br J Anaesth. 1979;51:431-434. carditis: diagnosis, antimicrobial therapy and management ment. J. Antimicrob Chemother. 2006;48:822-829.
425. Caputy AJ, Kim YI, Sanders DB. The neuromuscular of complications. Circulation. 2005;111:e394-e433. 478. Bratzler DW, Dellinger EP, Olsen KM, et al. Clinical prac-
blocking effects of therapeutic concentrations of various 452. Cosgrove SE, Vigliani GA, Campion M, et al. Initial low- tice guidelines for antimicrobial prophylaxis in surgery. Am
antibiotics on normal rat skeletal muscle: a quantitative dose gentamicin for Staphylococcus aureus bacteremia J Health Syst Pharm. 2013;70:195-283.
comparison. J Pharmacol Exp Ther. 1981;217:369-378. and endocarditis is nephrotoxic. Clin Infect Dis. 2009;48: 479. Nishimura RA, Carabello BA, Faxon DP, et al. ACC/AHA
426. Singh YN, Harvey AL, Marshall IG. Antibiotic-induced 713-721. 2008 guideline update on valvular heart disease: focused
paralysis of the mouse phrenic nerve-hemidiaphragm 453. Bayer AS, Murray BE. Initial low-dose aminoglycosides in update on infective endocarditis. A report of the American
preparation, and reversibility by calcium and by neostig- Staphylococcus aureus bacteremia: good science, urban College of Cardiology/American Heart Association Task
mine. Anesthesiology. 1978;48:418-424. legend, or just plain toxic? Clin Infect Dis. 2009;48: Force on Practice Guidelines; endorsed by the Society of
427. Proctor RA. Small colony variants: a pathogenic form of 722-724. Cardiovascular Anesthesiologists, Society for Cardiovascu-
bacteria that facilitates persistent and recurrent infections. 454. Bruss JB. Lack of evidence associating nephrotoxicity with lar Angiography and Interventions, and Society of Thoracic
Nat Rev Microbiol. 2006;4:295-305. low-dose gentamicin for Staphylococcus aureus bacteremia Surgeons. Circulation. 2008;118:887-896.
428. Vidal L, Gafter-Gvili A, Borok S, et al. Efficacy and safety and endocarditis. Clin Infect Dis. 2009;49:806. 480. Roos D, Dijksman LM, Oudemans-van Straaten HM, et al.
of aminoglycoside monotherapy: systematic review and 455. Hughes DW, Frei CR, Maxwell PR, et al. Continuous versus Randomized clinical trial of perioperative selective decon-
meta-analysis of randomized controlled trials. J Antimicrob intermittent infusion of oxacillin for treatment of infec- tamination of the digestive tract versus placebo in elective
Chemother. 2007;60:247-257. tious endocarditis caused by methicillin-susceptible Staph- gastrointestinal surgery. Brit J Surg. 2011;98:1365-1372.
429. Wilson WR, Karchmer AW, Dajani AS, et al. Antibiotic ylococcus aureus. Antimicrob Agents Chemother. 2009;53: 481. Saidel-Odes L, Polachek H, Peled N, et al. A randomized,
treatment of adults with infective endocarditis due to strep- 2014-2019. double-blind, placebo-controlled trial of selective digestive
tococci, enterococci, staphylococci and HACEK microor- 456. Gavalda J, Len O, Miro JM, et al. Treatment of Enterococcus decontamination using oral gentamicin and oral poly-
ganisms. JAMA. 1995;274:1706-1713. faecalis endocarditis with ampicillin plus ceftriaxone. Ann myxin E for eradication of carbapenem-resistant Klebsiella
430. Mylonakes E, Calderwood SB. Infective endocarditis in Intern Med. 2007;146:574-579. pneumoniae carriage. Infect Control Hosp Epidemiol. 2012;
adults. N Engl J Med. 2001;345:1318-1330. 457. Fernandez-Hidalgo N, Almirante B, Gavalda J, et al. Ampi- 33:14-19.
431. Solomkin JS, Mazuski JE, Bradley JS, et al. Diagnosis and cillin plus ceftriaxone is as effective as ampicillin plus gen- 482. Zuckerman T, Benyamini N, Sprecher H, et al. SCT in
management of complicated intra-abdominal infection in tamicin for treating Enterococcus faecalis infective patients with carbapenem resistant Klebsiella pneumoniae:
adults and children. Guidelines by the Surgical Infection endocarditis. Clin Infect Dis. 2013;56:1261-1268. a single center experience with oral gentamicin for the
Society and the Infectious Diseases Society of America. 458. Donowitz GR, Mandell GL. Empiric therapy for pneumo- eradication of carrier state. Bone Marrow Transplant. 2011;
Clin Infect Dis. 2010;50:133-164. nia. Rev Infect Dis. 1983;5(suppl 1):40-48. 46:1226-1230.
432. Hughes WT, Armstrong D, Bodey GP, et al. 2002 Guide- 459. Safdar N, Handelsman J, Maki DG. Does combination 483. Godbole G, Pai V, Kolvekar S, et al. Use of gentamicin-
lines for the use of antimicrobial agents in neutropenic intravenous antibiotic therapy reduce mortality in gram- collagen sponges in closure of sternal wounds in cardio­
patients with cancer. Clin Infect Dis. 2002;34:730-751. negative bacteremia? A meta-analysis. Lancet Infect Dis. thoracic surgery to reduce wound infections. Interact
433. Lew DP, Waldvogel FA. Osteomyelitis. N Engl J Med. 2004;4:519-527. Cardiovasc Thor Surg. 2012;14:390-394.
1997;336:999-1007. 460. Berlana D, Llop JM, Manresa F, et al. Outpatient treatment 484. Moran J, Sun S, Khababa I, et al. A randomized trial com-
434. Craig WA, Andes D. Aminoglycosides are useful for severe of Pseudomonas aeruginosa bronchial colonization with paring gentamicin/citrate and heparin locks for central
respiratory tract infections. Semin Respir Infect. 1997;12: long-term inhaled colistin, tobramycin, or both in adults venous catheters in maintenance hemodialysis patients.
271-277. without cystic fibrosis. Pharmacotherapy. 2011;31:146-157. Am J Kidney Dis. 2012;59:102-107.
435. Centers for Disease Control and Prevention. Sexually 461. Murray MP, Gowan JRW, Doherty CJ, et al. A randomized 485. Landry DL, Braden GL, Gobeille SL, et al. Emergence of
transmitted diseases, treatment guidelines, 2010. MMWR controlled trial of nebulized gentamicin in non-cystic gentamicin-resistant bacteremia in hemodialysis patients
Morb Mortal Wkly Rep. 2010;59(RR-6):1-116. fibrosis bronchiectasis. Am J Resp Crit Care Med. 2011;183: receiving gentamicin lock catheter prophylaxis. Clin J Am
436. Korvick JA, Bryan CS, Farber B, et al. Prospective observa- 491-499. Soc Nephrol. 2010;5:1799-1804.
tional study of Klebsiella bacteremia in 230 patients: 462. Ghannam DE, Rodrigues GH, Raad II, et al. Inhaled ami- 486. Chisholm SA, Quaye N, Cole MJ, et al. An evaluation
outcome for antibiotic combinations versus monotherapy. noglycosides in cancer patients with ventilator-associated of gentamicin susceptibility of Neisseria gonorrhoeae
Antimicrob Agents Chemother. 1992;36:2639-2644. gram-negative bacterial pneumonia: safety and feasibility isolates in Europe. J Antimicrob Chemother. 2011;66:
437. Neu HC. Tobramycin: an overview. J Infect Dis. 1976; in the era of escalating drug resistance. Eur J Clin Microbiol 592-595.
134(suppl):S3-S19. Infect Dis. 2009;28:253-259. 487. Josefsson G, Kolmert L. Prophylaxis with systematic anti-
438. Bendush L, Weber R. Tobramycin sulfate: a summary of 463. Wood GC. Aerosolized antibiotics for treating hospital- biotics versus gentamicin bone cement in total hip arthro-
worldwide experience from clinical trials. J Infect Dis. acquired and ventilator-associated pneumonia. Expert Rev plasty. A ten-year survey of 1,688 hips. Clin Orthopaed Rel
1976;134(suppl):S219-S234. Anti Infect Ther. 2011;9:993-1000. Res. 1993;292:210-214.
321.e7
488. Bertazzoni M, Benini E, Magnan A, et al. Release of gen- release, and biofilm formation. J Engineer Med. 2008;222: 495. van Kasteren ME, Mannien J, Oh A, et al. Antibiotic pro-
tamicin and vancomycin from temporary human hip 355-365. phylaxis and the risk of surgical site infections following
spacers in two stage revision of infected arthroplasty. 492. Hanssen AD, Spangehl MJ. Practical applications of total hip arthroplasty: timely administration is the most
J Antimicrob Chemother. 2004;53:329-334. antibiotic-loaded bone cement for treatment of infected important factor. Clin Infect Dis. 2007;44:921-927.

Chapter 25  Aminoglycosides


489. Bridgens J, Davies S, Tilley L, et al. Orthopedic bone joint replacements. Clin Orthopaed Rel Res. 2004;427: 496. Iarikov D, Damian H, Rubin D, et al. Choice and doses of
cement: do we know what we are using? J Bone Joint Surg 79-85. antibacterial agents for cement spacers in treatment of
Br. 2008;90:643-647. 493. Anagnostakos K, Hitzler P, Pape D, et al. Persistence of prosthetic joint infections: review of published studies. Clin
490. Postak PD, Greenwald AS. The influence of antibiotics on bacterial growth on antibiotic-loaded beads: is it actually a Infect Dis. 2012;55:1474-1480.
the fatigue life of acrylic bone cement. J Bone Joint Surg problem? Acta Orthop. 2008;79:302-307. 497. Barth RE, Voegely HC, Hoepelman AIM, et al. “To bead or
Am. 2006;88:148-155. 494. Dovas S, Liakopoulos V, Papatheodorou L, et al. Acute not to bead?” Treatment of osteomyelitis and prosthetic
491. Dunne NJ, Jill J, McAfee P, et al. Incorporation of large renal failure after antibiotic-impregnated bone cement joint-associated infections with gentamicin bead chains.
amounts of gentamicin sulphate into acrylic bone cement: treatment of an infected total knee arthroplasty. Clin Int J Antimicrob Agents. 2011;38:371-375.
effect on handling and mechanical properties, antibiotic Nephrol. 2008;69:207-212.

You might also like