You are on page 1of 19

44

C H A P T E R

Tetracyclines,
Macrolides, Clindamycin,
Chloramphenicol,
Streptogramins, &
Oxazolidinones
Camille E. Beauduy, PharmD, &
Lisa G. Winston, MD

C ASE STUDY

A 22-year-old woman presents to her college medical clinic motion tenderness is present. A first-catch urine specimen is
complaining of a 2-week history of vaginal discharge. She obtained for chlamydia and gonorrhea nucleic acid amplifi-
denies any fever or abdominal pain but does report vaginal cation testing. A urine pregnancy test is also ordered as the
bleeding after sexual intercourse. When questioned about patient reports she “missed her last period.” Pending these
her sexual activity, she reports having vaginal intercourse, results, the decision is made to treat her presumptively for
at times unprotected, with two men in the last 6 months. A chlamydial cervicitis. What are two potential treatment
pelvic examination is performed and is positive for muco- options for her possible chlamydial infection? How does her
purulent discharge from the endocervical canal. No cervical potential pregnancy affect the treatment decision?

The drugs described in this chapter inhibit bacterial protein OH O OH O


OH
synthesis by binding to and interfering with ribosomes. Most are 11 1 O
10 12 C
bacteriostatic, but a few are bactericidal against certain organ- 9 2
NH2
isms. Tetracycline and macrolide resistance is common. Except 8
7
3
OH
6 5 4
for tigecycline and the streptogramins, these antibiotics may be R6 OH H R5 H N(CH3)2
R7
administered orally. Renal
Clearance
R7 R6 R5 (mL/min)

■■ TETRACYCLINES
Chlortetracycline CI CH3 H 35
Oxytetracycline H CH3 OH 90
Tetracycline H CH3 H 65
Demeclocycline CI H H 35
All of the tetracyclines have the basic structure shown at right: Methacycline H CH2* OH 31
Doxycycline H CH3* OH 16
Minocycline N(CH3)2 H H 10
*There is no OH at position 6 on methacycline and doxycycline.

815
816    SECTION VIII  Chemotherapeutic Drugs

Free tetracyclines are crystalline amphoteric substances of low Tetracyclines are active against many Gram-positive and Gram-
solubility. They are available as hydrochlorides, which are more negative bacteria, including certain anaerobes, rickettsiae, chla-
soluble. Such solutions are acidic and fairly stable. Tetracyclines mydiae, and mycoplasmas. For susceptible organisms, differences
chelate divalent metal ions, which can interfere with their absorp- in clinical efficacy may be attributable to features of absorption,
tion and activity. Tigecycline is a glycylcycline and a semisynthetic distribution, and excretion of individual drugs. Tetracycline-
derivative of minocycline. resistant strains may be susceptible to doxycycline, minocycline,
and tigecycline, all of which are poor substrates for the efflux
Mechanism of Action & Antimicrobial pump, if that is the mechanism of resistance.
Activity
Tetracyclines are broad-spectrum bacteriostatic antibiotics that Resistance
inhibit protein synthesis. Tetracyclines enter microorganisms in Three mechanisms of resistance to tetracycline analogs have
part by passive diffusion and in part by an energy-dependent been described: (1) impaired influx or increased efflux by
process of active transport. Susceptible organisms concentrate an active transport protein pump; (2) ribosome protection
the drug intracellularly. Once inside the cell, tetracyclines bind due to production of proteins that interfere with tetracycline
reversibly to the 30S subunit of the bacterial ribosome, block- binding to the ribosome; and (3) enzymatic inactivation. The
ing the binding of aminoacyl-tRNA to the acceptor site on the most important of these are production of an efflux pump
mRNA-ribosome complex (Figure 44–1). This prevents addition and ribosomal protection. Tet(AE) efflux pump-expressing
of amino acids to the growing peptide. Gram-negative species are resistant to the older tetracyclines,

50S
ribosome

Amino acid
1 C
6
2
M
3

4
2 t6
5 6 1

Charged
tRNA
t5 4
t6
3

mRNA
30S
T

t5 Uncharged tRNA

FIGURE 44–1  Steps in bacterial protein synthesis and targets of several antibiotics. Amino acids are shown as numbered circles. The 70S
ribosomal mRNA complex is shown with its 50S and 30S subunits. In step 1, the charged tRNA unit carrying amino acid 6 binds to the acceptor
site on the 70S ribosome. The peptidyl tRNA at the donor site, with amino acids 1 through 5, then binds the growing amino acid chain to amino
acid 6 (peptide bond formation, step 2). The uncharged tRNA left at the donor site is released (step 3), and the new 6-amino acid chain with its
tRNA shifts to the peptidyl site (translocation, step 4). The antibiotic binding sites are shown schematically as triangles. Chloramphenicol (C) and
macrolides (M) bind to the 50S subunit and block peptide bond formation (step 2). The tetracyclines (T) bind to the 30S subunit and prevent
binding of the incoming charged tRNA unit (step 1).
CHAPTER 44  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol, Streptogramins, & Oxazolidinones     817

doxycycline, and minocycline. They are susceptible, how- Tetracyclines are classified as short-acting (tetracycline, as well
ever, to tigecycline, which is not a substrate of these pumps. as the agricultural agents chlortetracycline and oxytetracycline),
Similarly, a different pump [Tet(K)] of staphylococci confers intermediate-acting (demeclocycline), or long-acting (doxycy-
resistance to tetracycline, but not to doxycycline, minocycline, cline and minocycline) based on serum half-lives of 6–8 hours,
or tigecycline, none of which are pump substrates. The Tet(M) 12 hours, and 16–18 hours, respectively. Tigecycline has a half-life
ribosomal protection protein expressed by Gram-positives of 36 hours. The almost complete absorption and slow excretion
produces resistance to tetracycline, doxycycline, and mino- of doxycycline and minocycline allow for once-daily dosing for
cycline, but not to tigecycline, which, because of its bulky certain indications, but, by convention, these two drugs are usu-
t-butylglycylamido substituent, has a steric hindrance effect on ally dosed twice daily.
Tet(M) binding to the ribosome. Tigecycline is a substrate of
the chromosomally encoded multidrug efflux pumps of Proteus
sp and Pseudomonas aeruginosa, accounting for their intrinsic Clinical Uses
resistance to all tetracyclines including tigecycline. A tetracycline is the drug of choice in the treatment of most
infections caused by rickettsiae and Borrelia sp, including Rocky
Mountain spotted fever and Lyme disease. Tetracyclines are used
Pharmacokinetics preferentially to treat Anaplasma phagocytophilum and Ehrlichia
Tetracyclines differ in their absorption after oral administration sp. Tetracyclines are also excellent drugs for the treatment of
and in their elimination. Absorption after oral administration is Mycoplasma pneumoniae, chlamydiae, and some spirochetes. They
approximately 60–70% for tetracycline and demeclocycline (not are used in combination regimens to treat gastric and duodenal
typically used as an antibiotic; see below); and 95–100% for doxy- ulcer disease caused by Helicobacter pylori. They may be used in
cycline and minocycline. Tigecycline is poorly absorbed orally and various Gram-positive and Gram-negative bacterial infections,
must be administered intravenously. A portion of an orally admin- including vibrio infections, provided the organism is not resistant.
istered dose of tetracycline remains in the gut lumen, alters intes- In cholera, tetracyclines rapidly stop the shedding of vibrios, but
tinal flora, and is excreted in the feces. Absorption occurs mainly tetracycline resistance is an increasing problem. Tetracyclines
in the upper small intestine and is impaired by multivalent cations remain effective in most chlamydial infections, including sexually
(Ca2+, Mg2+, Fe2+, Al3+); by dairy products and antacids, which transmitted infections. Doxycycline is also an alternative agent
contain multivalent cations; and by alkaline pH. Tetracycline and recommended by the Centers for Disease Control and Preven-
demeclocycline should be administered on an empty stomach, tion for primary and secondary syphilis in patients with penicillin
while doxycycline and minocycline absorption is not impaired by allergy. A tetracycline—in combination with other antibiotics—is
food. Specially buffered doxycycline and minocycline solutions are indicated for plague, tularemia, and brucellosis. Tetracyclines are
formulated for intravenous administration. sometimes used in the treatment or prophylaxis of protozoal infec-
Tetracyclines are 40–80% bound by serum proteins. Oral tions, eg, those due to Plasmodium falciparum (see Chapter 52).
dosages of 500 mg every 6 hours of tetracycline hydrochlo- Other uses include treatment of acne, exacerbations of bronchitis,
ride produce peak blood levels of 4–6 mcg/mL. Peak levels of community-acquired pneumonia, leptospirosis, and some nontu-
2–4 mcg/mL are achieved with a 200-mg dose of doxycycline or berculous mycobacterial infections (eg, Mycobacterium marinum).
minocycline. Steady-state peak serum concentrations of tigecy- Tetracyclines formerly were used for a variety of common
cline are 0.6 mcg/mL at the standard dosage. Tetracyclines are dis- infections, including bacterial gastroenteritis and urinary tract
tributed widely to tissues and body fluids except for cerebrospinal infections. However, many strains of bacteria causing these infec-
fluid, where concentrations are 10–25% of those in serum. Tet- tions are now resistant, and other agents have largely supplanted
racyclines cross the placenta and are also excreted in breast milk. tetracyclines.
As a result of chelation with calcium, tetracyclines bind to—and Minocycline, 100 mg orally twice daily for 5 days, can eradi-
damage—growing bones and teeth. Carbamazepine, phenytoin, cate the meningococcal carrier state, but because of side effects
barbiturates, and chronic alcohol ingestion may shorten the half- and resistance of many meningococcal strains, ciprofloxacin or
life of tetracycline and doxycycline by 50% due to induction of rifampin is preferred. Demeclocycline is rarely used as an antibac-
hepatic enzymes that metabolize the drugs. terial, but it has been used off-label in the treatment of inappropri-
Tetracyclines are excreted mainly in bile and urine. Concen- ate secretion of antidiuretic hormone because of its inhibition of
trations in bile exceed those in serum tenfold. Some of the drug antidiuretic hormone in the renal tubule (see Chapter 15).
excreted in bile is reabsorbed from the intestine (enterohepatic Tigecycline, the first glycylcycline to reach clinical practice, has
circulation) and may contribute to maintenance of serum levels. several unique features that warrant its consideration apart from the
Ten to fifty percent of various tetracyclines is excreted into the older tetracyclines. Its spectrum is very broad, and many tetracy-
urine, mainly by glomerular filtration. Ten to forty percent of the cline-resistant strains are susceptible to tigecycline because it is not
drug is excreted in feces. Doxycycline and tigecycline, in contrast affected by the common resistance determinants. Susceptible organ-
to other tetracyclines, are eliminated by nonrenal mechanisms isms include coagulase-negative staphylococci and Staphylococcus
and do not accumulate significantly in renal failure, requiring no aureus, including methicillin-resistant, vancomycin-intermediate,
dosage adjustment. and vancomycin-resistant strains; streptococci, penicillin-susceptible
818    SECTION VIII  Chemotherapeutic Drugs

and resistant; enterococci, including vancomycin-resistant strains; Adverse Reactions


Gram-positive rods; Enterobacteriaceae; multidrug-resistant strains
Hypersensitivity reactions (drug fever, skin rashes) to tetracyclines
of Acinetobacter sp; anaerobes, both Gram-positive and Gram-
are uncommon. Most adverse effects are due to direct toxicity of
negative; rickettsiae, Chlamydia sp, and Legionella pneumophila;
the drug or to alteration of microbial flora.
and rapidly growing mycobacteria. Proteus and Providencia sp and
P aeruginosa, however, are intrinsically resistant.
A. Gastrointestinal Adverse Effects
Tigecycline, formulated for intravenous administration only,
is given as a 100-mg loading dose, then 50 mg every 12 hours. Nausea, vomiting, and diarrhea are the most common reasons for
As with all tetracyclines, tissue and intracellular penetration is discontinuing tetracyclines. These effects are attributable to direct
excellent; consequently, the volume of distribution is quite large local irritation of the intestinal tract. Oral tetracyclines can rarely
and peak serum concentrations are low. Elimination is primarily cause esophageal ulceration, so patients should be instructed to
biliary, and no dosage adjustment is needed for patients with renal take them with 8 ounces of water and remain upright for at least
insufficiency. In addition to the tetracycline class effects, the chief 30 minutes after each dose.
adverse effect of tigecycline is nausea, which occurs in up to one Tetracyclines alter the normal gastrointestinal flora, with sup-
third of patients, and occasionally vomiting. Neither nausea nor pression of susceptible coliform organisms and overgrowth of
vomiting usually requires discontinuation of the drug. Pseudomonas, Proteus, staphylococci, resistant coliforms, clostridia,
Tigecycline is approved for treatment of skin and skin- and Candida. This can result in intestinal functional disturbances,
structure infection, intra-abdominal infections, and community- anal pruritus, vaginal or oral candidiasis, or Clostridium difficile–
acquired pneumonia. However, in a meta-analysis of clinical trials, associated colitis. However, the risk of C difficile colitis may be
tigecycline was associated with a small but significant increase in lower with tetracyclines than with other antibiotics.
the risk of death compared with other antibiotics used to treat
these infections. The increased risk was most apparent in hospital- B. Bony Structures and Teeth
acquired and ventilator-associated pneumonia but was also seen in Tetracyclines are readily bound to calcium deposited in newly
other infections. This has led the U.S. Food and Drug Adminis- formed bone or teeth in young children. When a tetracycline
tration (FDA) to issue a black box warning that tigecycline should is given during pregnancy, it can be deposited in the fetal teeth,
be reserved for situations where alternative treatments are not suit- leading to fluorescence, discoloration, and enamel dysplasia. It
able. Because active drug concentrations in the urine and serum can also be deposited in bone, where it may cause deformity or
are relatively low, tigecycline may not be effective for urinary tract growth inhibition. Because of these effects, tetracyclines are gener-
infections or primary bacteremia. Tigecycline has in vitro activ- ally avoided in pregnancy. If the drug is given for long periods to
ity against a wide variety of multidrug-resistant pathogens (eg, children younger than 8 years, similar changes can result.
methicillin-resistant S aureus, extended-spectrum β-lactamase-
producing Gram-negatives, and Acinetobacter sp); however, its C. Other Toxicities
clinical efficacy in infections with multidrug-resistant organisms, Tetracyclines can impair hepatic function, especially during
compared with other agents, is unproven. pregnancy, in patients with preexisting liver disease, and when
high doses are given intravenously. Hepatic necrosis has been
A. Oral Dosage reported with daily doses of 4 g or more intravenously. Renal
tubular acidosis and Fanconi syndrome have been attributed to
The oral dosage for rapidly excreted tetracyclines, equivalent to
the administration of outdated tetracycline preparations. Tetracy-
tetracycline hydrochloride, is 0.25–0.5 g four times daily for
clines given along with diuretics may cause nephrotoxicity. Tetra-
adults and 25–50 mg/kg/d for children (8 years of age and older).
cycline and minocycline may accumulate to toxic levels in patients
For severe systemic infections, the higher dosage is indicated, at
with impaired kidney function. Intravenous injection can lead
least for the first few days. The dosage for doxycycline is 100 mg
to venous thrombosis. Intramuscular injection produces painful
once or twice daily; the minocycline dose is 100 mg twice daily.
local irritation and should be avoided. Systemically administered
Doxycycline is the oral tetracycline of choice for most indications
tetracyclines commonly induce sensitivity to sunlight or ultravio-
because it is generally well tolerated, it can be given twice daily,
let light, particularly in fair-skinned persons. Dizziness, vertigo,
and its absorption is not significantly affected by food. All tetracy-
and tinnitus have been noted, particularly with high doses or
clines chelate with metals, and none should be orally administered
prolonged administration of minocycline. These symptoms may
with milk, antacids, or ferrous sulfate. To avoid deposition in
also occur with higher doses of doxycycline.
growing bones or teeth, tetracyclines should be avoided in preg-
nant women and children younger than 8 years.

■■ MACROLIDES
B. Parenteral Dosage
Doxycycline and minocycline are available for intravenous injec- The macrolides are a group of closely related compounds charac-
tion at the same doses as the oral formulations. Intramuscular terized by a macrocyclic lactone ring (usually containing 14 or 16
injection is not recommended because of pain and inflammation atoms) to which deoxy sugars are attached. The prototype drug,
at the injection site. erythromycin, which consists of two sugar moieties attached to
CHAPTER 44  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol, Streptogramins, & Oxazolidinones     819

a 14-atom lactone ring, was obtained in 1952 from Streptomyces Resistance to erythromycin is usually plasmid-encoded. Three
erythreus, now called Saccharopolyspora erythraea. Clarithromycin general mechanisms have been identified: (1) reduced perme-
and azithromycin are semisynthetic derivatives of erythromycin. ability of the cell membrane or active efflux; (2) production
(by Enterobacteriaceae) of esterases that hydrolyze macrolides;
Macrolide O and (3) modification of the ribosomal binding site (so-called
ring
R1 R1 ribosomal protection) by chromosomal mutation or by a mac-
rolide-inducible or constitutive methylase. Efflux and methylase
OH
production are the most important resistance mechanisms in
R2 O R1
R1 6 OH
Gram-positive organisms. Cross-resistance is complete between
R1 R1 erythromycin and the other macrolides. Constitutive methylase
O
O O C2H5 production also confers resistance to structurally unrelated but
N(R1)2 mechanistically similar compounds such as clindamycin and strep-
Desosamine
O O togramin B (so-called macrolide-lincosamide-streptogramin, or
OH MLS-type B, resistance), which share the same ribosomal binding
R1
O site. Because nonmacrolides are poor inducers of the methylase,
HO R1 strains expressing an inducible methylase will appear susceptible
OR1
Cladinose in vitro. However, constitutive mutants that are resistant can be
R1 selected out and emerge during therapy with clindamycin.
Erythromycin (R1 = CH3, R2 = H)
Clarithromycin (R1, R2 = CH3) Pharmacokinetics
Erythromycin base is destroyed by stomach acid and must be
administered with enteric coating. Food interferes with absorp-
ERYTHROMYCIN
tion. The stearate and ethylsuccinate formulations are fairly acid-
resistant and somewhat better absorbed. A 500-mg intravenous
Chemistry dose of erythromycin lactobionate produces serum concentrations
The general structure of erythromycin is shown with the mac- of 10 mcg/mL 1 hour after dosing. The serum half-life is approxi-
rolide ring and the sugars desosamine and cladinose. It is poorly mately 1.5 hours normally and 5 hours in patients with anuria.
soluble in water (0.1%) but dissolves readily in organic solvents. Adjustment for renal failure is not necessary. Erythromycin is
Solutions are fairly stable at 4°C but lose activity rapidly at 20°C not removed by dialysis. Large amounts of an administered dose
and at acid pH. Erythromycins are usually dispensed as various are excreted in the bile, and only 5% is excreted in the urine.
esters and salts. Absorbed drug is distributed widely except to the brain and cere-
brospinal fluid. Erythromycin is taken up by polymorphonuclear
Mechanism of Action & Antimicrobial leukocytes and macrophages. It traverses the placenta and reaches
Activity the fetus.
The antibacterial action of erythromycin and other macrolides
may be inhibitory or bactericidal, particularly at higher concentra- Clinical Uses
tions, for susceptible organisms. Activity is enhanced at alkaline Erythromycin is a traditional drug of choice in corynebacterial
pH. Inhibition of protein synthesis occurs via binding to the 50S infections (diphtheria, corynebacterial sepsis, erythrasma) and
ribosomal RNA. The binding site is near the peptidyltransferase in respiratory, neonatal, ocular, or genital chlamydial infections.
center, and peptide chain elongation (ie, transpeptidation) is While it was used in treatment of community-acquired pneu-
prevented by blocking of the polypeptide exit tunnel. As a result, monia because its spectrum of activity includes pneumococcus,
peptidyl-tRNA is dissociated from the ribosome. Erythromy- M pneumoniae, and L pneumophila, newer macrolides are better
cin also inhibits the formation of the 50S ribosomal subunit tolerated and more commonly selected. Macrolide resistance is
(Figure 44–1). increasing in pneumococci and M pneumoniae. Erythromycin had
Erythromycin is active against susceptible strains of Gram-pos- also been useful as a penicillin substitute in penicillin-allergic indi-
itive organisms, especially pneumococci, streptococci, staphylo- viduals with infections caused by staphylococci and streptococci.
cocci, and corynebacteria. Mycoplasma pneumoniae, L pneumophila, Emergence of erythromycin resistance in staphylococci and in
Chlamydia trachomatis, Chlamydophila psittaci, Chlamydophila strains of group A streptococci has made macrolides less attractive
pneumoniae, H pylori, Listeria monocytogenes, and certain myco- as first-line agents for treatment of pharyngitis and skin and soft
bacteria (Mycobacterium kansasii, Mycobacterium scrofulaceum) tissue infections. Erythromycin has been studied as prophylaxis
also are susceptible. Gram-negative organisms such as Neisseria sp, against endocarditis during dental procedures in individuals with
Bordetella pertussis, Bartonella henselae, and Bartonella quintana as valvular heart disease, but clindamycin, which is better tolerated,
well as some Rickettsia species, Treponema pallidum, and Campylo- has largely replaced it.
bacter species are susceptible. Haemophilus influenzae is somewhat The oral dosage of erythromycin base or stearate is
less susceptible. 0.25–0.5 g every 6 hours (for children, 40 mg/kg/d). The dosage
820    SECTION VIII  Chemotherapeutic Drugs

of erythromycin ethylsuccinate is 0.4–0.8 g every 6 hours. Oral AZITHROMYCIN


erythromycin base (1 g) is sometimes combined with oral neo-
mycin or kanamycin for preoperative preparation of the colon. Azithromycin, a 15-atom lactone macrolide ring compound, is
The intravenous dosage of erythromycin lactobionate is 0.5–1.0 g derived from erythromycin by addition of a methylated nitrogen
every 6 hours for adults and 15–20 mg/kg/d divided every 6 hours into the lactone ring. Its spectrum of activity, mechanism of action,
for children. The higher dosage is recommended when treating and clinical uses are similar to those of clarithromycin. Azithromy-
pneumonia caused by L pneumophila. cin is active against M avium complex and T gondii. Azithromycin
is slightly less active than erythromycin and clarithromycin against
Adverse Reactions staphylococci and streptococci and slightly more active against
Anorexia, nausea, vomiting, and diarrhea are common. Gastro- H influenzae. Azithromycin is highly active against Chlamydia sp.
intestinal intolerance, which is due to a direct stimulation of gut Azithromycin differs from erythromycin and clarithromy-
motility, is the most common reason for selecting an alternative to cin mainly in pharmacokinetic properties. A 500-mg dose of
erythromycin. This side effect may actually be desirable in some azithromycin produces relatively low serum concentrations of
circumstances, leading to the off-label use of erythromycin to treat approximately 0.4 mcg/mL. However, azithromycin penetrates
patients with gastroparesis. into most tissues (except cerebrospinal fluid) and phagocytic
Erythromycins, particularly the older estolate formulation, can cells extremely well, with tissue concentrations exceeding serum
produce acute cholestatic hepatitis (fever, jaundice, impaired liver concentrations by 10- to 100-fold. The drug is slowly released
function), probably as a hypersensitivity reaction. Most patients from tissues (tissue half-life of 2–4 days) to produce an elimina-
recover from this, but hepatitis recurs if the drug is readministered. tion half-life approaching 3 days. These unique properties permit
Other allergic reactions include fever, eosinophilia, and rashes. once-daily dosing and shortening of the duration of treatment in
Erythromycin metabolites inhibit cytochrome P450 enzymes many cases. For example, a single 1-g dose of azithromycin is as
and, thus increase the serum concentrations of numerous drugs, effective as a 7-day course of doxycycline for chlamydial cervicitis
including theophylline, warfarin, cyclosporine, and methylpred- and urethritis. Azithromycin, as a 500-mg loading dose, followed
nisolone. Erythromycin increases serum concentrations of oral by a 250-mg single daily dose for the next 4 days, is commonly
digoxin by increasing its bioavailability. used alone or in combination with a beta-lactam antibiotic to treat
community-acquired pneumonia.
Azithromycin is rapidly absorbed and well tolerated orally. Alumi-
CLARITHROMYCIN num and magnesium antacids do not alter bioavailability but delay
absorption and reduce peak serum concentrations. Because it has a
Clarithromycin is derived from erythromycin by addition of a 15-member (not 14-member) lactone ring, azithromycin does not
methyl group and has improved acid stability and oral absorption inactivate cytochrome P450 enzymes and, therefore, is free of the
compared with erythromycin. Its mechanism of action is the same drug interactions that occur with erythromycin and clarithromycin.
as that of erythromycin. Clarithromycin and erythromycin are Macrolide antibiotics prolong the electrocardiographic QT
similar with respect to antibacterial activity except that clarithro- interval due to an effect on potassium ion channels. Prolongation
mycin is more active against Mycobacterium avium complex (see of the QT interval can lead to the torsades de pointes arrhythmia.
Chapter 47). Clarithromycin also has activity against Mycobacte- Recent studies have suggested that azithromycin may be associated
rium leprae, Toxoplasma gondii, and H influenzae. Erythromycin- with a small increased risk of cardiac death.
resistant streptococci and staphylococci are also resistant to
clarithromycin.
A 500-mg dose of clarithromycin produces serum concen- FIDAXOMICIN
trations of 2–3 mcg/mL. The longer half-life of clarithromycin
(6 hours) compared with erythromycin permits twice-daily Fidaxomicin, a minimally absorbed macrolide used to treat
dosing. The recommended dosage is 250–500 mg twice daily or Clostridium difficile infections, is discussed in Chapter 50.
1000 mg of the extended-release formulation once daily. Clar-
ithromycin penetrates most tissues well, with concentrations equal
to or exceeding serum concentrations. KETOLIDES
Clarithromycin is metabolized in the liver and is partially
eliminated in the urine. The major metabolite, 14-hydroxyclar- Ketolides are semisynthetic, 14-membered-ring macrolides, dif-
ithromycin, also has antibacterial activity and is eliminated in the fering from erythromycin by substitution of a 3-keto group for
urine. Dosage reduction (eg, a 500-mg loading dose, then 250 mg the neutral sugar l-cladinose. Telithromycin is approved for
once or twice daily) is recommended for patients with creatinine limited clinical use. It is active in vitro against Streptococcus pyo-
clearances less than 30 mL/min. Clarithromycin has drug interac- genes, S pneumoniae, S aureus, H influenzae, Moraxella catarrhalis,
tions similar to those described for erythromycin. Mycoplasma sp, L pneumophila, Chlamydia sp, H pylori, Neisseria
The advantages of clarithromycin compared with erythromy- gonorrhoeae, B fragilis, T gondii, and certain nontuberculous
cin are lower incidence of gastrointestinal intolerance and less mycobacteria. Many macrolide-resistant strains are susceptible to
frequent dosing. ketolides because the structural modification of these compounds
CHAPTER 44  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol, Streptogramins, & Oxazolidinones     821

renders them poor substrates for efflux pump–mediated resis- with aminoacyl translocation reactions. The binding site for
tance, and they bind to ribosomes of some bacterial species with clindamycin on the 50S subunit of the bacterial ribosome is
higher affinity than macrolides. identical with that for erythromycin. Streptococci, staphy-
Oral bioavailability of telithromycin is 57%, and tissue and lococci, and pneumococci are inhibited by clindamycin at
intracellular penetration is generally good. Telithromycin is a concentration of 0.5–5 mcg/mL. Enterococci and Gram-
metabolized in the liver and eliminated by a combination of negative aerobic organisms are resistant. Bacteroides sp and
biliary and urinary routes of excretion. It is administered as a other anaerobes are often susceptible, though resistance may
once-daily dose of 800 mg, which results in peak serum concen- be increasing, particularly in Gram-negative anaerobes. Resis-
trations of approximately 2 mcg/mL. It is a reversible inhibitor of tance to clindamycin, which generally confers cross-resistance
the CYP3A4 enzyme system and may slightly prolong the QTc to macrolides, is due to (1) mutation of the ribosomal recep-
interval. In the USA, telithromycin is now indicated only for tor site; (2) modification of the receptor by a constitutively
treatment of community-acquired bacterial pneumonia. Other expressed methylase (see section on erythromycin resistance,
respiratory tract infections were removed as indications when it above); and (3) enzymatic inactivation of clindamycin. Gram-
was recognized that use of telithromycin can result in hepatitis negative aerobic species are intrinsically resistant because of
and liver failure. Telithromycin is also contraindicated in patients poor permeability of the outer membrane.
with myasthenia gravis because it may exacerbate this condition.
Due to its potential for serious toxicity, an FDA-approved patient
medication guide detailing these risks must be dispensed to any Pharmacokinetics
patient receiving the medication. Oral dosages of clindamycin, 0.15–0.3 g every 8 hours
Solithromycin is a novel fluoroketolide that is pending FDA (10–20 mg/kg/d for children), yield serum levels of 2–3 mcg/mL.
approval after two phase 3 clinical trials showed noninferior- When administered intravenously, 600 mg of clindamycin every
ity when compared with moxifloxacin in the treatment of 8 hours gives levels of 5–15 mcg/mL. The drug is about 90%
community-acquired pneumonia. Although not yet marketed, protein-bound. Clindamycin penetrates well into most tissues,
the dose used in clinical trials was a loading dose of 800 mg with brain and cerebrospinal fluid being important exceptions.
orally or intravenously, followed by 400 mg daily for a total of It penetrates well into abscesses and is actively taken up and con-
5 days. The intravenous formulation was associated with higher centrated by phagocytic cells. Clindamycin is metabolized by the
rates of infusion-related reactions compared with moxifloxacin. liver, and both active drug and active metabolites are excreted in
Similar to telithromycin, solithromycin maintains in vitro activ- bile and urine. The half-life is about 2.5 hours in normal indi-
ity against macrolide-resistant bacteria, including S pneumoniae, viduals, increasing to 6 hours in patients with anuria. No dosage
staphylococci, enterococci, Chlamydia trachomatis, and Neisseria adjustment is required for renal failure.
gonorrhoeae. Its chemical structure lacks the pyridine-imidazole
side chain group, which is thought to contribute to telithromy-
cin’s hepatotoxicity; severe toxicity has not been demonstrated in Clinical Use
Phase II or III clinical trials. Clindamycin is indicated for the treatment of skin and soft-
tissue infections caused by streptococci and staphylococci. It
may be active against community-acquired strains of methi-
■■ CLINDAMYCIN cillin-resistant S aureus, though resistance has been increasing.
It is commonly used in conjunction with penicillin G to treat
Clindamycin is a chlorine-substituted derivative of lincomycin, toxic shock syndrome or necrotizing fasciitis caused by Group
an antibiotic that is elaborated by Streptomyces lincolnensis. A Streptococcus. In this setting, its use is typically limited to the
initial 48 to 72 hours of treatment with the goal of inhibiting
CH3
CH3 toxin production. Clindamycin is also indicated for treatment
N
CI CH
of infections caused by susceptible Bacteroides sp and other
C3H7 anaerobes. Clindamycin, sometimes in combination with an
C NH CH
aminoglycoside or cephalosporin, is used to treat penetrating
O
O HO wounds of the abdomen and the gut; infections originating in
OH the female genital tract, eg, septic abortion, pelvic abscesses, or
S CH3 pelvic inflammatory disease; and lung and periodontal abscesses.
OH Clindamycin is recommended for prophylaxis of endocarditis in
Clindamycin patients with specific valvular heart disease who are undergoing
certain dental procedures and have significant penicillin aller-
Mechanism of Action & Antibacterial gies. Clindamycin plus primaquine is an effective alternative
to trimethoprim-sulfamethoxazole for moderate to moderately
Activity severe Pneumocystis jiroveci pneumonia in AIDS patients. It is
Clindamycin, like erythromycin, inhibits protein synthesis also used in combination with pyrimethamine for AIDS-related
by interfering with the formation of initiation complexes and toxoplasmosis of the brain.
822    SECTION VIII  Chemotherapeutic Drugs

Adverse Effects ■■ CHLORAMPHENICOL


Common adverse effects are diarrhea, nausea, and skin rashes.
Impaired liver function (with or without jaundice) and neutro- Crystalline chloramphenicol is a neutral, stable compound with
penia sometimes occur. Administration of clindamycin is a risk the following structure:
factor for diarrhea and colitis due to C difficile. OH CH2OH O

NO2 C C N C CHCI2

■■ STREPTOGRAMINS H H H
Chloramphenicol

MECHANISM OF ACTION &


It is soluble in alcohol but poorly soluble in water. Chloram-
ANTIBACTERIAL ACTIVITY phenicol succinate, which is used for parenteral administration,
is highly water-soluble. It is hydrolyzed in vivo with liberation of
Quinupristin-dalfopristin is a combination of two
free chloramphenicol.
streptogramins—quinupristin, a streptogramin B, and dalfopris-
tin, a streptogramin A—in a 30:70 ratio. The streptogramins share
the same ribosomal binding site as the macrolides and clindamy- Mechanism of Action & Antimicrobial
cin and thus inhibit protein synthesis in an identical manner. Activity
Quinupristin-dalfopristin is rapidly bactericidal for most suscep-
Chloramphenicol is an inhibitor of microbial protein synthesis and
tible organisms except Enterococcus faecium, which is killed slowly.
is bacteriostatic against most susceptible organisms. It binds revers-
Quinupristin-dalfopristin is active against Gram-positive cocci,
ibly to the 50S subunit of the bacterial ribosome (Figure 44–1)
including multidrug-resistant strains of streptococci, penicillin-
and inhibits peptide bond formation (step 2). Chloramphenicol
resistant strains of S pneumoniae, methicillin-susceptible and resis-
is a broad-spectrum antibiotic that is active against both aerobic
tant strains of staphylococci, and E faecium (but not Enterococcus
and anaerobic Gram-positive and Gram-negative organisms. It is
faecalis). Resistance is due to modification of the quinupristin
active also against rickettsiae but not chlamydiae. Most Gram-
binding site (MLS-B type resistance), enzymatic inactivation of
positive bacteria are inhibited at concentrations of 1–10 mcg/mL,
dalfopristin, or efflux.
and many Gram-negative bacteria are inhibited by concentrations
of 0.2–5 mcg/mL. H influenzae, Neisseria meningitidis, and some
Pharmacokinetics strains of Bacteroides are highly susceptible; for these organisms,
Quinupristin-dalfopristin is administered intravenously at a chloramphenicol may be bactericidal.
dosage of 7.5 mg/kg every 8–12 hours. Peak serum concentra- Low-level resistance to chloramphenicol may emerge from
tions following an infusion of 7.5 mg/kg over 60 minutes are large populations of chloramphenicol-susceptible cells by selection
3 mcg/mL for quinupristin and 7 mcg/mL for dalfopristin. of mutants that are less permeable to the drug. Clinically signifi-
Quinupristin and dalfopristin are rapidly metabolized, with cant resistance is due to production of chloramphenicol acetyl-
half-lives of 0.85 and 0.7 hours, respectively. Elimination is prin- transferase, a plasmid-encoded enzyme that inactivates the drug.
cipally by the fecal route. Dose adjustment is not necessary for
renal failure, peritoneal dialysis, or hemodialysis. Patients with Pharmacokinetics
hepatic insufficiency may not tolerate the drug at usual doses,
however, because of increased area under the concentration The usual dosage of chloramphenicol is 50–100 mg/kg/d divided
curve of both parent drugs and metabolites. This may neces- every 6 hours. It is no longer available in the USA as an oral for-
sitate a dose reduction to 7.5 mg/kg every 12 hours or 5 mg/kg mulation. The parenteral formulation is a prodrug, chlorampheni-
every 8 hours. Quinupristin and dalfopristin significantly inhibit col succinate, which is hydrolyzed to yield free chloramphenicol,
CYP3A4, which metabolizes warfarin, diazepam, quetiapine, giving blood levels somewhat lower than those achieved with
simvastatin, and cyclosporine, among many others. Dosage orally administered drug. Chloramphenicol is widely distributed
reduction of cyclosporine may be necessary. to virtually all tissues and body fluids, including the central ner-
vous system and cerebrospinal fluid, such that the concentration
of chloramphenicol in brain tissue may be equal to that in serum.
Clinical Uses & Adverse Effects The drug penetrates cell membranes readily.
Quinupristin-dalfopristin is approved for treatment of infec- Most of the drug is inactivated either by conjugation with gluc-
tions caused by staphylococci or by vancomycin-resistant strains uronic acid (principally in the liver) or by reduction to inactive
of E faecium, but not E faecalis, which is intrinsically resistant, aryl amines. Active chloramphenicol, about 10% of the total dose
probably because of an efflux-type resistance mechanism. The administered, and its inactive degradation products are eliminated
principal toxicities are infusion-related events, such as pain at the in the urine. A small amount of active drug is excreted into bile
infusion site, and an arthralgia-myalgia syndrome. Quinupristin- and feces. There are no specific dosage adjustments recommended
dalfopristin is used to a limited extent in the USA due to the in renal or hepatic insufficiency; however, the drug will accumu-
availability of better-tolerated alternatives. late and should be used with extra caution in these situations.
CHAPTER 44  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol, Streptogramins, & Oxazolidinones     823

Newborns less than a week old and premature infants also clear Linezolid inhibits protein synthesis by preventing formation of the
chloramphenicol less well, and the dosage should be reduced to ribosome complex that initiates protein synthesis. Its unique binding
25 mg/kg/d. site, located on 23S ribosomal RNA of the 50S subunit, results in no
cross-resistance with other drug classes. Resistance is caused by muta-
Clinical Uses tion of the linezolid binding site on 23S ribosomal RNA.
Because of potential toxicity, bacterial resistance, and the availabil-
ity of many other effective alternatives, chloramphenicol is rarely Pharmacokinetics
used in the United States. It may be considered for treatment of Linezolid is 100% bioavailable after oral administration and has a
serious rickettsial infections such as typhus and Rocky Mountain half-life of 4–6 hours. It is metabolized by oxidative metabolism,
spotted fever. It is an alternative to a β-lactam antibiotic for treat- yielding two inactive metabolites. It is neither an inducer nor an
ment of bacterial meningitis occurring in patients who have major inhibitor of cytochrome P450 enzymes. Peak serum concentra-
hypersensitivity reactions to penicillin. tions average 18 mcg/mL following a 600-mg oral dose; cerebro-
spinal fluid (CSF) concentrations reach approximately 60–70% of
Adverse Reactions the serum level. The recommended dosage for most indications is
Adults occasionally develop gastrointestinal disturbances, includ- 600 mg twice daily, either orally or intravenously.
ing nausea, vomiting, and diarrhea. These symptoms are rare
in children. Oral or vaginal candidiasis may occur as a result of Clinical Uses
alteration of normal microbial flora. Linezolid is approved for vancomycin-resistant E faecium infec-
Chloramphenicol commonly causes a dose-related revers- tions, health care–associated pneumonia, community-acquired
ible suppression of red cell production at dosages exceeding pneumonia, and both complicated and uncomplicated skin and
50 mg/kg/d after 1–2 weeks. Aplastic anemia, a rare consequence soft tissue infections caused by susceptible Gram-positive bacte-
(1 in 24,000 to 40,000 courses of therapy) of chloramphenicol ria. Off-label uses of linezolid include treatment of multidrug-
administration by any route, is an idiosyncratic reaction unrelated resistant tuberculosis and Nocardia infections.
to dose, although it occurs more frequently with prolonged use.
Aplastic anemia tends to be irreversible and can be fatal, although
it may respond to bone marrow transplantation or immunosup- Adverse Effects
pressive therapy. Due to the severity of this reaction, a boxed The principal toxicity of linezolid is hematologic; the effects are
warning has been added to its U.S. labeling. reversible and generally mild. Thrombocytopenia is the most
Newborn infants lack an effective glucuronic acid conjugation common manifestation (seen in approximately 3% of treatment
mechanism for the degradation and detoxification of chloram- courses), particularly when the drug is administered for longer
phenicol. Consequently, when infants are given dosages above than 2 weeks. Anemia and neutropenia may also occur, most
50 mg/kg/d, the drug may accumulate, resulting in the gray baby commonly in patients with a predisposition to or underlying bone
syndrome, with vomiting, flaccidity, hypothermia, gray color, marrow suppression. Cases of optic and peripheral neuropathy
shock, and vascular collapse. To avoid this toxic effect, chloram- and lactic acidosis have been reported with prolonged courses of
phenicol should be used with caution in infants and the dosage linezolid. These side effects are thought to be related to linezolid-
limited to 50 mg/kg/d (or less during the first week of life) in full- induced inhibition of mitochondrial protein synthesis. There are
term infants and 25 mg/kg/d in premature infants. case reports of serotonin syndrome (see Chapter 16) occurring
Chloramphenicol inhibits hepatic microsomal enzymes that when linezolid is co-administered with serotonergic drugs, most
metabolize several drugs. Half-lives of these drugs are prolonged, frequently selective serotonin reuptake inhibitor antidepressants.
and the serum concentrations of phenytoin, tolbutamide, chlor- The FDA has issued a warning regarding the use of the drug with
propamide, and warfarin are increased. serotonergic agents.
Tedizolid is the active moiety of the prodrug tedizolid phos-
phate, a next-generation oxazolidinone, with high potency against
■■ OXAZOLIDINONES Gram-positive bacteria, including methicillin-resistant S aureus.
It is FDA-approved at a dose of 200 mg orally or intravenously
MECHANISM OF ACTION & once daily for 6 days for the treatment of skin and soft tissue
ANTIMICROBIAL ACTIVITY infection. Potential advantages over linezolid include increased
potency against staphylococci and a longer half-life of 12 hours,
Linezolid is a member of the oxazolidinone class of synthetic allowing once-daily dosing. It may be associated with a decreased
antimicrobials. It is active against Gram-positive organisms risk of marrow suppression; however, it has not been studied over
including staphylococci, streptococci, enterococci, Gram-positive a prolonged duration of therapy. It is thought to have a lower risk
anaerobic cocci, and Gram-positive rods such as corynebacteria, of serotonergic toxicity, but concomitant use with serotonin reup-
Nocardia sp, and L monocytogenes. It is primarily a bacteriostatic take inhibitors has not been formally evaluated. Tedizolid is more
agent but is bactericidal against streptococci. It is also active highly protein-bound (70–90%) than linezolid (31%); there are
against Mycobacterium tuberculosis. no data on CSF penetration of tedizolid.
824    SECTION VIII  Chemotherapeutic Drugs

SUMMARY  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol,


Streptogramins, & Oxazolidinones
Pharmacokinetics, Toxicities,
Subclass, Drug Mechanism of Action Effects Clinical Applications Interactions

TETRACYCLINES
  •  Tetracycline Prevents bacterial protein Bacteriostatic activity Infections caused by Oral • mixed clearance (half-life 8 h) • dosed
synthesis by binding to the against susceptible mycoplasma, chlamydiae, every 6 h • divalent cations impair oral
30S ribosomal subunit bacteria rickettsiae, some spirochetes absorption • Toxicity: Gastrointestinal upset,
• malaria • H pylori • acne hepatotoxicity, photosensitivity, deposition in
bone and teeth

  • Doxycycline: Oral and IV; longer half-life (18 h) so dosed twice daily; nonrenal elimination; absorption is minimally affected by divalent cations; used to treat community-
acquired pneumonia and exacerbations of bronchitis
  •  Minocycline: Oral and IV; longer half-life (16 h) so dosed twice daily; frequently causes reversible vestibular toxicity
  •  Tigecycline: IV; unaffected by common tetracycline resistance mechanisms; very broad spectrum of activity against Gram-positive, Gram-negative, and anaerobic
bacteria; nausea and vomiting are the primary toxicities

MACROLIDES
  •  Erythromycin Prevents bacterial protein Bacteriostatic activity Community-acquired Oral, IV • hepatic clearance (half-life 1.5 h)
synthesis by binding to the against susceptible pneumonia • pertussis • dosed every 6 h • cytochrome P450 inhibitor
50S ribosomal subunit bacteria • corynebacterial and • Toxicity: Gastrointestinal upset, hepatotoxicity,
chlamydial infections QTc prolongation

  •  Clarithromycin: Oral; longer half-life (6 h) so dosed twice daily; added activity versus M avium complex, toxoplasma, and M leprae
  • Azithromycin: Oral, IV; very long half-life (68 h) allows for once-daily dosing and 5-day course of therapy of community-acquired pneumonia; does not inhibit cytochrome
P450 enzymes
  • Telithromycin: Oral; unaffected by efflux-mediated resistance so is active versus many erythromycin-resistant strains of pneumococci; rare cases of fulminant hepatic
failure

LINCOSAMIDE
  •  Clindamycin Prevents bacterial protein Bacteriostatic activity Skin and soft tissue infections Oral, IV • hepatic clearance (half-life 2.5 h)
synthesis by binding to the against susceptible • anaerobic infections • dosed every 6–8 hours • Toxicity:
50S ribosomal subunit bacteria Gastrointestinal upset, C difficile colitis

STREPTOGRAMINS
  • Quinupristin- Prevents bacterial protein Rapid bactericidal Infections caused by IV • hepatic clearance • dosed every 8–12 h
dalfopristin synthesis by binding to the activity against most staphylococci or vancomycin- • cytochrome P450 inhibitor • Toxicity: Severe
50S ribosomal subunit susceptible bacteria resistant strains of E faecium infusion-related myalgias and arthralgias

CHLORAMPHENICOL
  Prevents bacterial protein Bacteriostatic activity Use is rare in the developed IV • hepatic clearance (half-life 2.5 h) • dosage is
synthesis by binding to the against susceptible world because of serious 50–100 mg/kg/d in four divided doses
50S ribosomal subunit bacteria toxicities • Toxicity: Dose-related anemia, idiosyncratic
aplastic anemia, gray baby syndrome

OXAZOLIDINONES
  •  Linezolid Prevents bacterial protein Bacteriostatic activity Infections caused by Oral, IV • hepatic clearance (half-life 6 h) • dosed
synthesis by binding to the against susceptible methicillin-resistant twice-daily • Toxicity: Duration-dependent bone
23S ribosomal RNA of 50S bacteria staphylococci and vancomycin- marrow suppression, neuropathy, and optic
subunit resistant enterococci neuritis • serotonin syndrome may occur when
co-administered with other serotonergic drugs
(eg, selective serotonin reuptake inhibitors)

Tedizolid: Oral and IV; longer half-life (12 h) so dosed once daily; increased potency versus staphylococci; approved for use in skin and soft tissue infections.
CHAPTER 44  Tetracyclines, Macrolides, Clindamycin, Chloramphenicol, Streptogramins, & Oxazolidinones     825

P R E P A R A T I O N S Chopra I, Roberts M: Tetracycline antibiotics: Mode of action, applications,


molecular biology, and epidemiology of bacterial resistance. Microbiol Mol
A V A I L A B L E Biol Rev 2001;65:232.
De Vriese AS et al: Linezolid-induced inhibition of mitochondrial protein synthe-
sis. Clin Infect Dis 2006;42:1111.
GENERIC NAME AVAILABLE AS
Dryden MS: Linezolid pharmacokinetics and pharmacodynamics in clinical treat-
Chloramphenicol Generic, Chloromycetin ment. 2011;66(Suppl 4):S7.
TETRACYCLINES File Jr. TM et al: SOLITAIRE-IV: A randomized, double-blind, multicenter study
Demeclocycline Generic, Declomycin comparing the efficacy and safety of intravenous-to-oral solithromycin to
intravenous-to-oral moxifloxacin for treatment of community-acquired
Doxycycline Generic, Vibramycin, others bacterial pneumonia. Clin Infect Dis 2016;63:1007.
Minocycline Generic, Minocin, others Hancock RE: Mechanisms of action of newer antibiotics for gram-positive
Tetracycline Generic, others pathogens. Lancet Infect Dis 2005;5:209.
Tigecycline Tygacil Leclerq R: Mechanisms of resistance to macrolides and lincosamides: Nature
of the resistance elements and their clinical implications. Clin Infect Dis
MACROLIDES
2002;34:482.
Azithromycin Generic, Zithromax Lee M et al: Linezolid for treatment of chronic extensively drug-resistant tubercu-
Clarithromycin Generic, Biaxin losis. N Engl J Med 2012;367:1508.
Erythromycin Generic, others Livermore DM: Tigecycline: What is it, and where should it be used? J Antimicrob
KETOLIDES Chemother 2005;56:611.
Moran GJ et al: Methicillin-resistant S aureus infections among patients in the
Telithromycin Ketek
emergency department. N Engl J Med 2006;355:666.
LINCOMYCIN Moran GJ et al: Tedizolid for 6 days versus linezolid for 10 days for acute bacterial
Clindamycin Generic, Cleocin skin and skin-structure infections (ESTABLISH-2): A randomized, double-
STREPTOGRAMINS blind, phase 3, non-inferiority trial. Lancet 2014;14:696.
Quinupristin and dalfopristin Synercid Prokocimer P et al: Tedizolid phosphate vs linezolid for treatment of acute bacterial
skin and skin structure infections. JAMA 2013;309:559.
OXAZOLIDINONE
Tasina E et al: Efficacy and safety of tigecycline for the treatment of infectious
Linezolid Generic, Zyvox diseases: A meta-analysis. Lancet Infect Dis 2011;11:834.
Tedizolid Sivextro Van Bambeke F: Renaissance of antibiotics against difficult infections: Focus on
oritavancin and new ketolides and quinolones. Ann Med 2014;46:512.
Wayne RA et al: Azithromycin and risk of cardiovascular death. N Engl J Med
2012;366:1881.
Woytowish MR, Rowe AS: Clinical relevance of linezolid-associated serotonin
toxicity. Ann Pharmacother 2013;47:388.
REFERENCES Zuckerman JM: Macrolides and ketolides: Azithromycin, clarithromycin, telithro-
Barrera CM et al: Efficacy and safety of oral solithromycin versus oral moxifloxa- mycin. Infect Dis Clin North Am 2004;18:621.
cin for treatment of community-acquired bacterial pneumonia: A global,
double-blind, multicenter, randomized, active-controlled, non-inferiority
trial (SOLITAIRE-ORAL). Lancet 2016;16:421.

C ASE STUDY ANSWER

A tetracycline or a macrolide is effective in the treatment of patient is pregnant, then tetracyclines would be contraindi-
chlamydial cervicitis. Doxycycline at a dose of 100 mg PO cated and she should receive azithromycin, which is safe in
bid for 7 days is the preferred tetracycline, while azithro- pregnancy.
mycin as a single 1 g dose is the preferred macrolide. If the
45
C H A P T E R

Aminoglycosides &
Spectinomycin
Camille E. Beauduy, PharmD, & Lisa G. Winston, MD*

C ASE STUDY

A 45-year-old man with no significant medical history was pending. The ICU attending physician is concerned about a
admitted to the intensive care unit (ICU) 10 days ago after bloodstream infection and decides to treat with empiric com-
suffering third-degree burns over 40% of his body. He had bination therapy directed against Pseudomonas aeruginosa.
been relatively stable until the last 24 hours. Now, he is febrile The combination therapy includes tobramycin. The patient
(39.5°C [103.1°F]), and his white blood cell count has risen weighs 70 kg (154 lb) and has an estimated creatinine clear-
from 8500 to 20,000/mm3. He has also had an episode of hypo- ance of 90 mL/min. How should tobramycin be dosed using
tension (86/50 mmHg) that responded to a fluid bolus. Blood once-daily and conventional dosing strategies? How should
cultures were obtained at the time of his fever and results are each regimen be monitored for efficacy and toxicity?

The drugs described in this chapter are bactericidal inhibitors of pro- which various amino sugars are attached by glycosidic linkages
tein synthesis that interfere with ribosomal function. These agents (Figures 45–1 and 45–2). They are water-soluble, stable in solu-
are useful mainly against aerobic Gram-negative microorganisms. tion, and more active at alkaline than at acid pH.

B. Mechanism of Action
■■ AMINOGLYCOSIDES The mode of action of streptomycin has been studied more
closely than that of other aminoglycosides, but all aminoglyco-
The aminoglycosides include streptomycin, neomycin, kanamy- sides are thought to act similarly. Aminoglycosides are irrevers-
cin, amikacin, gentamicin, tobramycin, sisomicin, netilmicin, ible inhibitors of protein synthesis, but the precise mechanism
and others. They are used most widely in combination with other for bactericidal activity is unclear. The initial event is passive dif-
agents to treat drug-resistant organisms; for example, they are fusion via porin channels across the outer membrane (see Figure
used with a β-lactam antibiotic in serious infections with Gram- 43–3). Drug is then actively transported across the cell mem-
negative bacteria, with a β-lactam antibiotic or vancomycin for brane into the cytoplasm by an oxygen-dependent process. The
Gram-positive endocarditis, and with one or more agents for treat- transmembrane electrochemical gradient supplies the energy for
ment of mycobacterial infections, such as tuberculosis. this process, and transport is coupled to a proton pump. Low
extracellular pH and anaerobic conditions inhibit transport by
General Properties of Aminoglycosides reducing the gradient. Transport may be enhanced by cell wall-
A. Physical and Chemical Properties active drugs such as penicillin or vancomycin; this enhancement
Aminoglycosides have a hexose ring, either streptidine (in strep- may be the basis of the synergism of those antibiotics with
tomycin) or 2-deoxystreptamine (in other aminoglycosides), to aminoglycosides.
Inside the cell, aminoglycosides bind to 30S-subunit ribosomal
*
The authors thank Drs. Henry F. Chambers and Daniel H. Deck for proteins. Protein synthesis is inhibited by aminoglycosides in at
their contributions to previous editions. least three ways (Figure 45–3): (1) interference with the initiation

826
CHAPTER 45  Aminoglycosides & Spectinomycin    827

complex of peptide formation; (2) misreading of mRNA, which


NH2 causes incorporation of incorrect amino acids into the peptide and
C NH results in a nonfunctional protein; and (3) breakup of polysomes
CH3
into nonfunctional monosomes. These activities occur more or
HO NH CH2OH
NH O
CHO
O
less simultaneously, and the overall effect is irreversible and leads
OH
H2N C NH O O OH
to cell death.

HO OH NH OH C. Mechanisms of Resistance
Three principal mechanisms of resistance have been established:
CH3
(1) production of a transferase enzyme that inactivates the amino-
Streptidine Streptose N-methyl-L- glycoside by adenylylation, acetylation, or phosphorylation. This
glucosamine
is the principal type of resistance encountered clinically. (2) There
Streptobiosamine is impaired entry of aminoglycoside into the cell. This may result
from mutation or deletion of a porin protein involved in transport
FIGURE 45–1  Structure of streptomycin. and maintenance of the electrochemical gradient or from growth

1 2
H2C NH2 NH2 H2C NH2 NH2
O O
5 3 2 NH R NH2
HO 4 I 1 O 4 II 1 HO I O II
3 2 5 6 CH2 OH CH2 OH
O O
HO OH HO O 5 NH2 HO O
4 3 1 III 4 OH III OH
2 3

HO NH2 HO NH2
5 Tobramycin

Kanamycin R =H
O OH NH2
HO
Amikacin R = C CH CH2 CH2 NH2 NH O

I O II NH R
R1

HC NH R2 NH2
NH2 HO O O
O OH
5 3 2 NH R3 Plazomicin
I II III
4 O 1
O OH
CH3
O R C CH CH2 CH2 NH2
NH2 HO O OH HO NH–CH3
III
2 CH3

HO NH CH3
Gentamicin, netilmicin

Ring I Ring II

C4–C5
R1 R2 bond R3
Gentamicin C1 CH3 CH3 Single H
Gentamicin C2 CH3 H Single H
Gentamicin C1a H H Single H
Netilmicin H H Double C 2H 5

FIGURE 45–2  Structures of several important aminoglycoside antibiotics. Ring II is 2-deoxystreptamine. The resemblance between kana-
mycin and amikacin and between gentamicin, netilmicin, and tobramycin can be seen. Plazomicin’s ring II and III are similar to the other struc-
tures; it shares the same hydroxyl-aminobutyric acid R group as amikacin. Its ring I differs from amikacin in that it is unsaturated. The circled
numerals on the kanamycin molecule indicate points of attack of plasmid-mediated bacterial transferase enzymes that can inactivate this drug.
➀, ➁, and ➂, acetyltransferase; ➃, phosphotransferase; ➄, adenylyltransferase. Amikacin is resistant to modification at ➁,➂,➃, and ➄; whereas
plazomicin is resistant to modification at ➀, ➁, ➃, and ➄.
828    SECTION VIII  Chemotherapeutic Drugs

Normal bacterial cell

Initiation 50S subunit Nascent peptide chain


codon

30S subunit
mRNA 3´

Aminoglycoside-treated bacterial cell

Drug (block of Drug (miscoded peptide chain)


initiation complex)
Drug (block of

translocation)

30S subunit
mRNA 3´

FIGURE 45–3  Putative mechanisms of action of the aminoglycosides in bacteria. Normal protein synthesis is shown in the top panel. At
least three aminoglycoside effects have been described, as shown in the bottom panel: block of formation of the initiation complex; miscoding
of amino acids in the emerging peptide chain due to misreading of the mRNA; and block of translocation on mRNA. Block of movement of the
ribosome may occur after the formation of a single initiation complex, resulting in an mRNA chain with only a single ribosome on it, a so-called
monosome. (Reproduced, with permission, from Trevor AT, Katzung BG, Masters SB: Pharmacology: Examination & Board Review, 6th ed. McGraw-Hill, 2002. Copyright © The
McGraw-Hill Companies, Inc.)

conditions under which the oxygen-dependent transport process bile, the level may reach 30% of that in blood. With prolonged
is not functional. (3) The receptor protein on the 30S ribosomal therapy, diffusion into pleural or synovial fluid may result in con-
subunit may be deleted or altered as a result of a mutation. centrations 50–90% of that of plasma.
Traditionally, aminoglycosides have been administered in two
D. Pharmacokinetics and Once-Daily Dosing or three equally divided doses per day in patients with normal
Aminoglycosides are absorbed very poorly from the intact gastro- renal function. However, administration of the entire daily dose
intestinal tract, and almost the entire oral dose is excreted in feces in a single injection may be preferred in many clinical situations
after oral administration. However, the drugs may be absorbed if for at least two reasons. Aminoglycosides exhibit concentration-
ulcerations are present. Aminoglycosides are usually administered dependent killing; that is, higher concentrations kill a larger pro-
intravenously as a 30–60 minute infusion. After intramuscular portion of bacteria and kill at a more rapid rate. They also have a
injection, aminoglycosides are well absorbed, giving peak concen- significant postantibiotic effect, such that the antibacterial activity
trations in blood within 30–90 minutes. After a brief distribution persists beyond the time during which measurable drug is present.
phase, peak serum concentrations are identical to those following The postantibiotic effect of aminoglycosides can last several hours.
intravenous injection. The normal half-life of aminoglycosides Because of these properties, a given total amount of aminoglycoside
in serum is 2–3 hours, increasing to 24–48 hours in patients may have better efficacy when administered as a single large dose
with significant impairment of renal function. Aminoglycosides than when administered as multiple smaller doses.
are only partially and irregularly removed by hemodialysis—eg, When administered with a cell wall-active antibiotic (a β-lactam
40–60% for gentamicin—and even less effectively by peritoneal or vancomycin), aminoglycosides may exhibit synergistic killing
dialysis. Aminoglycosides are highly polar compounds that do not against certain bacteria. The effect of the drugs in combination
enter cells readily. They are largely excluded from the central ner- is greater than the anticipated effect of each individual drug; ie,
vous system and the eye. In the presence of active inflammation, the killing effect of the combination is more than additive. This
however, cerebrospinal fluid levels reach 20% of plasma levels, synergy may be important in certain clinical situations, such as
and, in neonatal meningitis, the levels may be higher. Intrathecal endocarditis.
or intraventricular injection is required for high levels in cerebro- Adverse effects from aminoglycosides are both time- and
spinal fluid. Even after parenteral administration, concentrations concentration-dependent. Toxicity is unlikely to occur until a
of aminoglycosides are not high in most tissues except the renal certain threshold concentration is reached, but, once that concentra-
cortex. Concentration in most secretions is also modest; in the tion is achieved, the time beyond this threshold becomes critical.
CHAPTER 45  Aminoglycosides & Spectinomycin    829

This threshold is not precisely defined, but a trough concentration regimen, peak serum concentrations should be determined from
above 2 mcg/mL is predictive of toxicity. At clinically relevant a blood sample obtained 30–60 minutes after a dose, and trough
doses, the total time above this threshold is greater with multiple concentrations from a sample obtained just before the next dose.
smaller doses of drug than with a single large dose. Doses of gentamicin and tobramycin should be adjusted to main-
Numerous clinical studies demonstrate that a single daily dose tain peak levels between 5 and 10 mcg/mL (typically between
of aminoglycoside is just as effective—and probably less toxic— 8 and 10 mcg/mL in more serious infections) and trough levels
than multiple smaller doses. Therefore, many authorities recom- <2 mcg/mL (<1 mcg/mL is optimal).
mend that aminoglycosides be administered as a single daily dose
in most clinical situations. However, the efficacy of once-daily E. Adverse Effects
aminoglycoside dosing in combination therapy of enterococcal All aminoglycosides are ototoxic and nephrotoxic. Ototoxicity and
and staphylococcal endocarditis in patients with a prosthetic valve nephrotoxicity are more likely to be encountered when therapy is
remains to be defined, and administration of lower doses two or continued for more than 5 days, at higher doses, in the elderly,
three times daily is still recommended. In contrast, limited data do and in the setting of renal insufficiency. Concurrent use with loop
support once-daily dosing in streptococcal endocarditis. The role diuretics (eg, furosemide, ethacrynic acid) or other nephrotoxic
of once-daily dosing in pregnancy, obesity, and in neonates also is antimicrobial agents (eg, vancomycin or amphotericin) can poten-
not well defined. tiate nephrotoxicity and should be avoided if possible. Ototoxicity
Once-daily dosing has potential practical advantages. For can manifest either as auditory damage, resulting in tinnitus and
example, repeated determinations of serum concentrations are high-frequency hearing loss initially, or as vestibular damage with
unnecessary unless an aminoglycoside is given for more than 3 vertigo, ataxia, and loss of balance. Nephrotoxicity results in rising
days. A drug administered once a day rather than three times a serum creatinine levels or reduced creatinine clearance, although
day is less labor intensive. And once-a-day dosing is more feasible the earliest indication often is an increase in trough serum ami-
for outpatient therapy. noglycoside concentrations. Neomycin, kanamycin, and amikacin
Aminoglycosides are cleared by the kidney, and excretion is are the agents most likely to cause auditory damage. Streptomycin
directly proportional to creatinine clearance. To avoid accumu- and gentamicin are the most vestibulotoxic. Neomycin, tobramy-
lation and toxic levels, once-daily dosing of aminoglycosides is cin, and gentamicin are the most nephrotoxic.
generally avoided if renal function is impaired. Rapidly changing In very high doses, aminoglycosides can produce a curare-like
renal function, which may occur with acute kidney injury, must effect with neuromuscular blockade that results in respiratory
also be monitored to avoid overdosing or underdosing. Provided paralysis. This paralysis is usually reversible by calcium gluconate,
these pitfalls are avoided, once-daily aminoglycoside dosing is safe when given promptly, or neostigmine. Hypersensitivity occurs
and effective. If the creatinine clearance is >60 mL/min, then a infrequently.
single daily dose of 5–7 mg/kg of gentamicin or tobramycin is
recommended (15 mg/kg for amikacin). For patients with cre- F. Clinical Uses
atinine clearance <60 mL/min, traditional dosing as described Aminoglycosides are mostly used against aerobic Gram-negative
below is recommended. With once-daily dosing, serum concen- bacteria, especially when there is concern for drug-resistant patho-
trations need not be routinely checked until the second or third gens or in critically ill patients. They are almost always used in
day of therapy, depending on the stability of renal function and combination with a β-lactam antibiotic to extend empiric cover-
the anticipated duration of therapy. In most circumstances, it is age and to take advantage of the potential synergism between these
unnecessary to check peak concentrations; an exception may be two classes of drugs. Penicillin-aminoglycoside combinations have
when ensuring adequately high peak concentrations for treat- also been used to achieve bactericidal activity in treatment of
ing infections caused by drug-resistant pathogens. The goal is to enterococcal endocarditis and to shorten duration of therapy for
administer drug so that concentrations of <1 mcg/mL are present viridans streptococcal endocarditis. Due to toxicity, these com-
between 18 and 24 hours after dosing. This provides sufficient binations are used less frequently when alternate regimens are
time for washout of drug to occur before the next dose is given. available. For example, in the case of enterococcal endocarditis,
Several nomograms have been developed and validated to assist studies suggest that the combination of ampicillin and ceftriaxone
clinicians with once-daily dosing (eg, Freeman reference). is an effective regimen with less risk for nephrotoxicity. When
With traditional dosing, adjustments must be made to pre- aminoglycosides are used, the selection of agent and dose depends
vent accumulation of drug and toxicity in patients with renal on the infection being treated and the susceptibility of the isolate.
insufficiency. Either the dose of drug is kept constant and the
interval between doses is increased, or the interval is kept con-
stant and the dose is reduced. Nomograms and formulas have STREPTOMYCIN
been constructed relating serum creatinine levels to adjust-
ments in traditional treatment regimens. Because aminoglycoside Streptomycin (Figure 45–1) was isolated from a strain of Strepto-
clearance is directly proportional to the creatinine clearance, a myces griseus. The antimicrobial activity of streptomycin is typical
method for determining the aminoglycoside dose is to estimate of that of other aminoglycosides, as are the mechanisms of resis-
creatinine clearance using the Cockcroft-Gault formula described tance. Resistance has emerged in most species, restricting the cur-
in Chapter 60. For a traditional twice- or thrice-daily dosing rent usefulness of streptomycin, with the exceptions listed below.
830    SECTION VIII  Chemotherapeutic Drugs

Ribosomal resistance to streptomycin develops readily, limiting its Resistance


role as a single agent.
Streptococci and enterococci are relatively resistant to gentamicin
owing to failure of the drug to penetrate into the cell. However,
Clinical Uses gentamicin in combination with some penicillins or vancomycin
A. Mycobacterial Infections produces a potent bactericidal effect, which in part is due to
Streptomycin is mainly used as a second-line agent for treatment enhanced uptake of drug that occurs with inhibition of cell wall
of tuberculosis. The dosage is 15 mg/kg/d with a maximum synthesis. Resistance to gentamicin rapidly emerges in staphy-
of 1 g/d (20–40 mg/kg/d for children), and it may be given lococci during monotherapy owing to selection of permeability
intramuscularly or intravenously. It should be used only in com- mutants. Ribosomal resistance is rare. Among Gram-negative
bination with other agents to prevent emergence of resistance. bacteria, resistance is most commonly due to plasmid-encoded
See Chapter 47 for additional information regarding the use of aminoglycoside-modifying enzymes. Gram-negative bacteria that
streptomycin in mycobacterial infections. are gentamicin-resistant usually are susceptible to amikacin, which
is much more resistant to modifying enzyme activity. The entero-
B. Nontuberculous Infections coccal enzyme that modifies gentamicin is a bifunctional enzyme
In plague, tularemia, and sometimes, brucellosis, streptomycin, that also inactivates amikacin, netilmicin, and tobramycin but not
1 g twice daily (15 mg/kg twice daily for children), is given intra- streptomycin; the latter is modified by a different enzyme. This
muscularly in combination with an oral tetracycline. is why some gentamicin-resistant enterococci are susceptible to
Penicillin plus streptomycin is effective for enterococcal endo- streptomycin.
carditis and 2-week therapy of viridans streptococcal endocarditis;
however, for susceptible strains, gentamicin is used more commonly Clinical Uses
when an aminoglycoside is selected as adjunct therapy. Streptomy- A. Intramuscular or Intravenous Administration
cin remains a useful agent for treating gentamicin non-susceptible Gentamicin is used mainly in severe infections caused by Gram-
enterococcal infections, as some isolates that are resistant to gen- negative bacteria that are likely to be resistant to other drugs,
tamicin (and therefore resistant to netilmicin, tobramycin, and especially P aeruginosa, Enterobacter sp, Serratia marcescens,
amikacin) will remain susceptible to streptomycin. Proteus sp, Acinetobacter sp, and Klebsiella sp. It usually is used
in combination with a second agent because an aminoglycoside
Adverse Reactions alone may not be effective for infections outside the urinary tract.
Fever, skin rashes, and other allergic manifestations may result Aminoglycosides should not be used as single agents for therapy
from hypersensitivity to streptomycin. This occurs most frequently of pneumonia because penetration of infected lung tissue is poor
with a prolonged course of treatment (eg, for tuberculosis). and local conditions of low pH and low oxygen tension contribute
Pain at the injection site is common but usually not severe. to limited activity. Gentamicin 5–7 mg/kg/d traditionally is given
The most serious toxic effect with streptomycin is disturbance of intravenously in three equal doses, but once-daily administration
vestibular function—vertigo and loss of balance. The frequency is just as effective for some organisms and less toxic (see above).
and severity of this disturbance are in proportion to the age of Gentamicin, in combination with a cell wall-active antibiotic,
the patient, the blood levels of the drug, and the duration of may also be indicated in the treatment of endocarditis caused by
administration. Vestibular dysfunction may follow a few weeks of Gram-positive bacteria (streptococci, staphylococci, and entero-
unusually high blood levels (eg, in individuals with impaired renal cocci) as discussed earlier.
function) or months of relatively low blood levels. Vestibular tox-
icity tends to be irreversible. Streptomycin given during pregnancy B. Topical and Ocular Administration
can cause deafness in the newborn. Creams, ointments, and solutions containing 0.1–0.3% genta-
micin sulfate have been used for the treatment of infected burns,
wounds, or skin lesions and in attempts to prevent intravenous
GENTAMICIN catheter infections. The effectiveness of topical preparations for
these indications is unclear. Topical gentamicin is partly inacti-
Gentamicin is a mixture of three closely related constituents, vated by purulent exudates. Gentamicin can be injected intraocu-
C1, C1A, and C2 (Figure 45–2) isolated from Micromonospora larly for treatment of certain eye infections.
purpurea. It is effective against both Gram-positive and Gram-
negative organisms, and many of its properties resemble those of C. Intrathecal Administration
other aminoglycosides.
Meningitis caused by Gram-negative bacteria has been treated
by the intrathecal injection of gentamicin sulfate, 1–10 mg/d.
Antimicrobial Activity However, neither intrathecal nor intraventricular gentamicin was
Gentamicin sulfate, 2–10 mcg/mL, inhibits in vitro many beneficial in neonates with meningitis, and intraventricular gen-
strains of staphylococci and Gram-negative bacteria, including tamicin was toxic, raising questions about the usefulness of this
P. aeruginosa and Enterobacteriaceae. Like all aminoglycosides, it form of therapy. Moreover, the availability of third-generation
has no activity against anaerobes. cephalosporins for Gram-negative meningitis has rendered this
CHAPTER 45  Aminoglycosides & Spectinomycin    831

therapy obsolete in most cases. It may be used in cases of drug- AMIKACIN


resistant meningitis or severe β-lactam allergy.
Amikacin is a semisynthetic derivative of kanamycin; it is less
Adverse Reactions toxic than the parent molecule (Figure 45–2). It is resistant to
many enzymes that inactivate gentamicin and tobramycin, and,
Nephrotoxicity is usually reversible upon drug discontinuation. It
therefore, can be used against some microorganisms resistant to
occurs in 5–25% of patients receiving gentamicin for longer than
the latter drugs. Many Gram-negative bacteria, including many
3–5 days. Such toxicity requires, at the very least, adjustment of the
strains of Proteus, Pseudomonas, Enterobacter, and Serratia, are
dosing regimen and should prompt reconsideration of the need for the
inhibited by 1–20 mcg/mL amikacin in vitro. After injection of
drug, particularly if there is a less toxic alternative agent. Measurement
500 mg of amikacin every 12 hours (15 mg/kg/d) intramuscularly,
of gentamicin serum levels is essential. Ototoxicity, which tends to be
peak levels in serum are 10–30 mcg/mL.
irreversible, manifests itself mainly as vestibular dysfunction. Loss of
Strains of multidrug-resistant Mycobacterium tuberculosis,
hearing can also occur. Ototoxicity is in part genetically determined,
including streptomycin-resistant strains, are usually susceptible
having been linked to point mutations in mitochondrial DNA, and
to amikacin. Kanamycin-resistant strains may be cross-resis-
occurs in 1–5% for patients receiving gentamicin for more than
tant to amikacin. The dosage of amikacin for tuberculosis is
5 days. Hypersensitivity reactions to gentamicin are uncommon.
10–15 mg/kg/d as a once-daily or two to three times weekly
injection and always in combination with other drugs to which
TOBRAMYCIN the isolate is susceptible.
Like all aminoglycosides, amikacin is nephrotoxic and ototoxic
This aminoglycoside (Figure 45–2) has an antibacterial spectrum (particularly for the auditory portion of the eighth nerve). Serum
similar to that of gentamicin. Although there is some cross- concentrations should be monitored. Target peak serum concen-
resistance between gentamicin and tobramycin, it is unpredictable trations for an every-12-hours dosing regimen are 20–40 mcg/mL,
in individual strains. Separate laboratory susceptibility tests are and trough levels should be maintained between 4 and 8 mcg/mL.
therefore necessary.

A. Intramuscular or Intravenous Administration NETILMICIN


The pharmacokinetic properties of tobramycin are virtually iden-
Netilmicin shares many characteristics with gentamicin and tobra-
tical with those of gentamicin. The daily dose of tobramycin is
mycin. However, the addition of an ethyl group to the 1-amino
5–7 mg/kg intramuscularly or intravenously, traditionally divided
position of the 2-deoxystreptamine ring (ring II, Figure 45–2)
into three equal amounts and given every 8 hours but now often
sterically protects the netilmicin molecule from enzymatic degra-
given as a single daily dose. Monitoring blood levels in renal insuf-
dation at the 3-amino (ring II) and 2-hydroxyl (ring III) positions.
ficiency is an essential guide to proper dosing.
Consequently, netilmicin may be active against some gentamicin-
Tobramycin has almost the same antibacterial spectrum as gen-
resistant and tobramycin-resistant bacteria.
tamicin with a few exceptions. Gentamicin is slightly more active
The dosage (5–7 mg/kg/d) and the routes of administration are
against S marcescens, whereas tobramycin is slightly more active against
the same as for gentamicin. Netilmicin is largely interchangeable
P aeruginosa; Enterococcus faecalis is susceptible to both gentamicin and
with gentamicin or tobramycin but is no longer available in the
tobramycin, but E faecium is resistant to tobramycin. Gentamicin and
United States.
tobramycin are otherwise interchangeable clinically.
Like other aminoglycosides, tobramycin is ototoxic and neph-
rotoxic. Nephrotoxicity of tobramycin may be slightly less than NEOMYCIN, KANAMYCIN, &
that of gentamicin.
PAROMOMYCIN
B. Inhaled and Ophthalmic Administration
Neomycin, kanamycin, and paromomycin have similar pharma-
Tobramycin is formulated in solution (300 mg in 5 mL) for inha- cologic properties.
lation for treatment of P aeruginosa lower respiratory tract infec-
tions complicating cystic fibrosis. The drug is recommended as a
300-mg dose regardless of the patient’s age or weight for admin- Antimicrobial Activity & Resistance
istration twice daily in repeated cycles of 28 days on therapy, fol- Drugs of the neomycin group are active against Gram-positive and
lowed by 28 days off therapy. Serum concentrations 1 hour after Gram-negative bacteria and some mycobacteria. P aeruginosa and
inhalation average 1 mcg/mL; consequently, nephrotoxicity and streptococci are generally resistant. Mechanisms of antibacterial
ototoxicity rarely occur. Caution should be used when adminis- action and resistance are the same as with other aminoglycosides.
tering tobramycin to patients with preexisting renal, vestibular, or The former widespread use of these drugs in bowel preparation for
hearing disorders. Tobramycin is also available as 0.3% ophthal- elective surgery contributed to the selection of resistant organisms
mic ointment and drops for the treatment of superficial eye infec- and some outbreaks of enterocolitis in hospitals. Cross-resistance
tions. These formulations result in minimal systemic absorption between kanamycin and neomycin is complete and may result in
and are unlikely to cause systemic adverse effects. amikacin cross-resistance.
832    SECTION VIII  Chemotherapeutic Drugs

Pharmacokinetics vestibular function. Deafness may occur, especially in adults with


impaired renal function and prolonged elevation of drug levels.
Like all aminoglycosides, drugs of the neomycin group are poorly
The sudden absorption of postoperatively instilled kanamycin
absorbed from the gastrointestinal tract. After oral administra-
from the peritoneal cavity (3–5 g) has resulted in curare-like neu-
tion, the intestinal flora is suppressed or modified, and the drug
romuscular blockade and respiratory arrest. Calcium gluconate
is excreted in the feces. Excretion of any absorbed drug is mainly
and neostigmine can act as antidotes.
through glomerular filtration into the urine.
Although hypersensitivity is not common, prolonged appli-
cation of neomycin-containing ointments to skin and eyes has
Clinical Uses resulted in severe allergic reactions.
Neomycin is generally limited to topical and oral use due to toxicity
associated with parenteral use and higher resistance rates compared
to other aminoglycosides. Kanamycin use is limited to treatment of PLAZOMICIN
multi-drug-resistant tuberculosis, although alternate agents, such as
Plazomicin is a new aminoglycoside under development and is
amikacin, may be preferred. It is no longer available in the USA.
expected to undergo review by the U.S. Food and Drug Admin-
Paromomycin has been shown to be effective against visceral leish-
istration in 2017. It has been studied in phase II clinical trials for
maniasis when given parenterally (see Chapter 52), and this serious
treatment of urinary tract infections; phase III clinical trials are
infection may represent an important use for this drug. Paromomy-
underway for treatment of carbapenem-resistant Enterobacteria-
cin can be used for intestinal Entamoeba histolytica infection and is
ceae. It is a synthetic molecule derived from sisomicin, an ami-
sometimes used for intestinal infections with other parasites.
noglycoside no longer available,. Various structural modifications
have yielded a compound less susceptible to most aminoglycoside
A. Topical Administration
modifying enzymes, thus retaining activity against aminogly-
Solutions containing 1–5 mg/mL neomycin have been used on coside-resistant pathogens. It appears to have similarly potent
infected surfaces or injected into joints, the pleural cavity, tissue in vitro activity against Enterobacteriaceae and displays two- to
spaces, or abscess cavities where infection is present. The total four-fold lower MICs against nonfermenting Gram-negative
amount of drug given in this fashion must be limited to 15 mg/kg/d bacilli (eg, P aeruginosa) when compared with gentamicin, tobra-
because at higher doses enough drug may be absorbed to produce mycin, and amikacin. It has activity similar to gentamicin against
systemic toxicity. Whether topical application for active infection staphylococci. A 15-mg/kg dose yields mean peak and trough con-
adds anything to appropriate systemic therapy is questionable. centrations of 113 mcg/mL and 0.43 mcg/mL, respectively. The
Ointments, often formulated as a neomycin-polymyxin-bacitracin half-life is about 4 hours, and it is being studied as a single daily
combination, can be applied to infected skin lesions or in the nares dose. Due to limited clinical experience, it is unclear whether the
for suppression of staphylococci but they are largely ineffective. toxicity profile will be similar to other aminoglycosides; however,
no ototoxicity or nephrotoxicity has been observed in early trials.
B. Oral Administration
In preparation for elective bowel surgery, 1 g of neomycin may
be given orally every 6–8 hours for 1–2 days, often combined ■■ SPECTINOMYCIN
with 1 g of erythromycin base. This reduces the aerobic bowel
flora with little effect on anaerobes. In hepatic encephalopathy, Spectinomycin is an aminocyclitol antibiotic that is structurally
coliform flora can be suppressed by giving 1 g every 6–8 hours related to aminoglycosides. It lacks amino sugars and glycosidic
together with reduced protein intake, thus reducing ammonia bonds.
production. Use of neomycin for hepatic encephalopathy has been CH3 OH
largely supplanted by lactulose and other medications that are less O O
HN CH3
toxic. Use of paromomycin in the treatment of protozoal infec-
tions is discussed in Chapter 52.
HO
O OH
C. Intravenous and Intramuscular Administration NH O
When used intravenously, the standard dose for kanamycin is
CH3
15 mg/kg/day in two to three divided doses, whereas for treatment
Spectinomycin
of tuberculosis, 15 mg/kg is usually given intramuscularly as a sin-
gle daily dose. In the case of once daily administration, kanamycin
Spectinomycin is active in vitro against many Gram-positive
peak concentrations are typically between 35 and 45 mcg/mL,
and Gram-negative organisms, but it is used almost solely as
while trough concentrations should be undetectable.
an alternative treatment for drug-resistant gonorrhea or gonor-
rhea in penicillin-allergic patients. The majority of gonococcal
Adverse Reactions isolates are inhibited by 6 mcg/mL of spectinomycin. Strains of
All members of the neomycin group have significant nephro- gonococci may be resistant to spectinomycin, but there is no
toxicity and ototoxicity. Auditory function is affected more than cross-resistance with other drugs used in gonorrhea. Notably, it is
CHAPTER 45  Aminoglycosides & Spectinomycin    833

not recommended for treatment of pharyngeal gonococcal infec- children).There is pain at the injection site and, occasionally, fever
tions due to high failure rates regardless of in vitro susceptibility. and nausea. Nephrotoxicity and anemia have been observed rarely.
Spectinomycin is rapidly absorbed after intramuscular injection. Spectinomycin is no longer available for use in the USA but is still
The standard regimen is a single dose of 2–4 g/d (40 mg/kg in recommended elsewhere.

SUMMARY Aminoglycosides
Subclass, Mechanism of Pharmacokinetics, Toxicities,
Drug Action Effects Clinical Applications Interactions

AMINOGLYCOSIDES & SPECTINOMYCIN


  •  Gentamicin Prevents bacterial Bactericidal activity against Sepsis caused by aerobic IV • renal clearance (half-life 2.5 h) • conventional
protein synthesis susceptible bacteria • synergistic Gram-negative bacteria dosing 1.3–1.7 mg/kg q8h with goal peak levels
by binding to the effects against Gram-positive bacteria • synergistic activity in 5–8 mcg/mL • trough levels <2 mcg/mL
30S ribosomal when combined with β-lactams or endocarditis caused by • once-daily dosing at 5–7 mg/kg as effective and
subunit vancomycin • concentration- streptococci, staphylococci, may have less toxicity than conventional dosing
dependent killing and a significant and enterococci • Toxicity: Nephrotoxicity (reversible), ototoxicity
post-antibiotic effect (irreversible), neuromuscular blockade

  •  Tobramycin: Intravenous; more active than gentamicin versus Pseudomonas; may also have less nephrotoxicity
  • Amikacin: Intravenous; resistant to many enzymes that inactivate gentamicin and tobramycin; higher doses and target peaks and troughs than gentamicin and
tobramycin
  •  Streptomycin: Intramuscular, widespread resistance limits use to specific indications such as tuberculosis and enterococcal endocarditis
  •  Neomycin: Oral or topical, poor bioavailability; used before bowel surgery to decrease aerobic flora
  • Spectinomycin: Intramuscular; sole use is for treatment of antibiotic-resistant gonococcal infections or gonococcal infections in penicillin-allergic patients; not available
in the USA

P R E P A R A T I O N S Cheer SM, Waugh J, Noble S: Inhaled tobramycin (TOBI): A review of its use in
the management of Pseudomonas aeruginosa infections in patients with cystic
A V A I L A B L E fibrosis. Drugs 2003;63:2501.
Freeman CD et al: Once-daily dosing of aminoglycosides: Review and recommen-
dations for clinical practice. J Antimicrob Chemother 1997;39:677.
GENERIC NAME AVAILABLE AS
Jackson J et al: Aminoglycosides: How should we use them in the 21st century?
Amikacin Generic, Amikin Curr Opin Infect Dis 2013;26:516.
Gentamicin Generic Le T, Bayer AS: Combination antibiotic therapy for infective endocarditis. Clin
Kanamycin Generic, Kantrex Infect Dis 2003;36:615.
Neomycin Generic, Mycifradin Olsen KM et al: Effect of once-daily dosing vs. multiple daily dosing of tobramycin
on enzyme markers of nephrotoxicity. Crit Care Med 2004;32:1678.
Paromomycin Generic, Humatin
Paul M et al: Beta-lactam monotherapy versus beta-lactam-aminoglycoside combi-
Streptomycin Generic nation therapy in cancer patients with neutropenia. Cochrane Database Syst
Tobramycin Generic, Nebcin Rev 2013 Jun 29;6:CD003038.
Peña C et al: Effect of adequate single-drug versus combination antimicrobial
therapy on mortality in Pseudomonas aeruginosa bloodstream infections. Clin
REFERENCES Infect Dis 2013;57:208.
Baddour L et al: Infective Endocarditis in Adults: Diagnosis, Antimicrobial Poole K: Aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob Agents
Therapy, and Management of Complications. Circulation 2015;132:1435. Chemother 2005;49:479.
Busse H-J, Wöstmann C, Bakker EP: The bactericidal action of streptomycin: Zhanel G et al: Comparison of the next generation aminoglycoside plazomicin
Membrane permeabilization caused by the insertion of mistranslated pro- to gentamicin, tobramycin, and amikacin. Expert Rev Anti Infect Ther
teins into the cytoplasmic membrane of Escherichia coli and subsequent 2012;10:459.
caging of the antibiotic inside the cells due to degradation of these proteins.
J Gen Microbiol 1992;138:551.

C ASE STUDY ANSWER

The patient has normal renal function and thus qualifies divided and administered every 8 hours, as a conventional
for once-daily dosing. Tobramycin could be administered dosing strategy. With conventional dosing, peak and trough
as a single once-daily injection at a dose of 350–490 mg concentrations should be monitored with the target peak
(5–7 mg/kg). A serum level between 1.5 and 6 mcg/mL mea- concentration of 5–10 mcg/mL and the target trough con-
sured 8 hours after infusion correlates with an appropriate centration of <2 mcg/mL.
trough level. Alternatively, the same total daily dose could be

You might also like