Download as pdf or txt
Download as pdf or txt
You are on page 1of 71

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
43
C H A P T E R

Beta-Lactam & Other Cell


Wall- & Membrane-Active
Antibiotics
Camille E. Beauduy, PharmD, & Lisa G. Winston, MD*

C ASE STUDY

A 45-year-old man is brought to the local hospital emer- (38.7°C [101.7°F]), hypotensive (90/54 mmHg), tachypneic
gency department by ambulance. His wife reports that (36/min), and tachycardic (110/min). He has no signs of
he had been in his normal state of health until 3 days ago meningismus but is oriented only to person. A stat chest
when he developed a fever and a productive cough. Dur- x-ray shows a left lower lung consolidation consistent with
ing the last 24 hours he has complained of a headache and pneumonia. A CT scan is not concerning for lesions or
is increasingly confused. His wife reports that his medical elevated intracranial pressure. The plan is to start empiric
history is significant only for hypertension, for which he antibiotics and perform a lumbar puncture to rule out
takes hydrochlorothiazide and lisinopril, and that he is bacterial meningitis. What antibiotic regimen should be
allergic to amoxicillin. She says that he developed a rash prescribed to treat both pneumonia and meningitis? Does
many years ago when prescribed amoxicillin for bron- the history of amoxicillin rash affect the antibiotic choice?
chitis. In the emergency department, the man is febrile Why or why not?

■■ BETA-LACTAM COMPOUNDS Structural integrity of the 6-aminopenicillanic acid nucleus (rings


A plus B) is essential for the biologic activity of these compounds.
Hydrolysis of the β-lactam ring by bacterial β-lactamases yields
PENICILLINS penicilloic acid, which lacks antibacterial activity.
The penicillins share features of chemistry, mechanism of action,
A. Classification
pharmacology, and immunologic characteristics with cephalospo-
rins, monobactams, carbapenems, and β-lactamase inhibitors. Substituents of the 6-aminopenicillanic acid moiety determine the
All are β-lactam compounds, so named because of their four- essential pharmacologic and antibacterial properties of the result-
membered lactam ring. ing molecules. Penicillins can be assigned to one of three groups
(below). Within each of these groups are compounds that are
relatively stable to gastric acid and suitable for oral administration,
Chemistry eg, penicillin V, dicloxacillin, and amoxicillin. The side chains of
All penicillins have the basic structure shown in Figure 43–1. A some representatives of each group are shown in Figure 43–2.
thiazolidine ring (A) is attached to a β-lactam ring (B) that car-
ries a secondary amino group (RNH–). Substituents (R; examples 1. Penicillins (eg, penicillin G)—These have greatest activ-
shown in Figure 43–2) can be attached to the amino group. ity against Gram-positive organisms, Gram-negative cocci, and
non-β-lactamase-producing anaerobes. However, they have little
*
The authors thank Dr. Henry F. Chambers and Dr. Daniel Deck for activity against Gram-negative rods, and they are susceptible to
their contributions to this chapter in previous editions. hydrolysis by β-lactamases.

795
796    SECTION VIII  Chemotherapeutic Drugs

H H H H
Amidase S
S CH3
CH3
R N C C C R N CH CH C
CH3 Penicillin CH3
B A H
C N C C N CH COOH
O COOH
Lactamase O
Substituted 6-aminopenicillanic acid
6-Aminopenicillanic acid
The following structures can each be substituted
O H H H
at the R to produce a new penicillin.
S H
R1 C N C C C R
B A H
Cephalosporin
C N C
O C CH2 R2 CH2 Penicillin G
COOH
Substituted 7-aminocephalosporanic acid

OCH2 Penicillin V
O H H H

R C N C C CH3
B Monobactam
C N
C C Oxacillin
O SO3H
N C
Substituted 3-amino-4-methylmonobactamic acid O CH3
(aztreonam)
Cl

HO H H C C Dicloxacillin
HC C C N C
B S R Cl O CH3
H3C
C N
Carbapenem
O COOH
OC2H5
NH
Nafcillin
R: CH2 CH2 NH CH

Substituted 3-hydroxyethylcarbapenemic acid


(imipenem)

CH Ampicillin
FIGURE 43–1  Core structures of four β-lactam antibiotic
NH2
families. The ring marked B in each structure is the β-lactam ring.
The penicillins are susceptible to inactivation by amidases and
lactamases at the points shown. Note that the carbapenems have HO CH Amoxicillin
a different stereochemical configuration in the lactam ring that
imparts resistance to most common β-lactamases. Substituents for NH2
the penicillin and cephalosporin families are shown in Figures 43–2
and 43–6, respectively.
CH

NHCO
2. Antistaphylococcal penicillins (eg, nafcillin)—These Piperacillin
penicillins are resistant to staphylococcal β-lactamases. They N O
are active against staphylococci and streptococci but not against
enterococci, anaerobic bacteria, and Gram-negative cocci and
rods. O
N
3. Extended-spectrum penicillins (aminopenicillins and C2H5
antipseudomonal penicillins)—These drugs retain the anti-
bacterial spectrum of penicillin and have improved activity against
FIGURE 43–2  Side chains of some penicillins (R groups).
Gram-negative rods. Like penicillin, however, they are relatively
susceptible to hydrolysis by β-lactamases.
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     797

B. Penicillin Units and Formulations polysaccharides and peptides known as peptidoglycan. The poly-
The activity of penicillin G was originally defined in units. Crys- saccharide contains alternating amino sugars, N-acetylglucosamine
talline sodium penicillin G contains approximately 1600 units and N-acetylmuramic acid (Figure 43–4). A five-amino-acid
per mg (1 unit = 0.6 mcg; 1 million units of penicillin = 0.6 g). peptide is linked to the N-acetylmuramic acid sugar. This peptide
Semisynthetic penicillins are prescribed by weight rather than terminates in d-alanyl-d-alanine. Penicillin-binding protein (PBP,
units. The minimum inhibitory concentration (MIC) of any an enzyme) removes the terminal alanine in the process of forming
penicillin (or other antimicrobial) is usually given in mcg/mL. a cross-link with a nearby peptide. Cross-links give the cell wall its
Most penicillins are formulated as the sodium or potassium salt of rigidity. Beta-lactam antibiotics, structural analogs of the natural
the free acid. Potassium penicillin G contains about 1.7 mEq of d-Ala-d-Ala substrate, covalently bind to the active site of PBPs.
K+ per million units of penicillin (2.8 mEq/g). Nafcillin contains This binding inhibits the transpeptidation reaction (Figure 43–5)
Na+, 2.8 mEq/g. Procaine salts and benzathine salts of penicillin G and halts peptidoglycan synthesis, and the cell dies. The exact
provide repository forms for intramuscular injection. In dry crys- mechanism of cell death is not completely understood, but auto-
talline form, penicillin salts are stable for years at 4°C. Solutions lysins are involved in addition to the disruption of cross linking of
lose their activity rapidly (eg, within 24 hours at 20°C) and must the cell wall. Beta-lactam antibiotics kill bacterial cells only when
be prepared fresh for administration. they are actively growing and synthesizing cell wall.

Mechanism of Action Resistance


Penicillins, like all β-lactam antibiotics, inhibit bacterial growth Resistance to penicillins and other β-lactams is due to one of four
by interfering with the transpeptidation reaction of bacterial cell general mechanisms: (1) inactivation of antibiotic by β-lactamase,
wall synthesis. The cell wall is a rigid outer layer that completely (2) modification of target PBPs, (3) impaired penetration of drug
surrounds the cytoplasmic membrane (Figure 43–3), maintains to target PBPs, and (4) antibiotic efflux. Beta-lactamase produc-
cell integrity, and prevents cell lysis from high osmotic pressure. tion is the most common mechanism of resistance. Hundreds
The cell wall is composed of a complex, cross-linked polymer of of different β-lactamases have been identified. Some, such as

Porin

Outer
membrane

Cell
wall

Peptidoglycan

β Lactamase
Periplasmic
space

PBP PBP

Cytoplasmic
membrane

FIGURE 43–3  A highly simplified diagram of the cell envelope of a Gram-negative bacterium. The outer membrane, a lipid bilayer, is
present in Gram-negative but not Gram-positive organisms. It is penetrated by porins, proteins that form channels providing hydrophilic
access to the cytoplasmic membrane. The peptidoglycan layer is unique to bacteria and is much thicker in Gram-positive organisms than in
Gram-negative ones. Together, the outer membrane and the peptidoglycan layer constitute the cell wall. Penicillin-binding proteins (PBPs) are
membrane proteins that cross-link peptidoglycan. Beta-lactamases, if present, reside in the periplasmic space or on the outer surface of the
cytoplasmic membrane, where they may destroy β-lactam antibiotics that penetrate the outer membrane.
798    SECTION VIII  Chemotherapeutic Drugs

those produced by Staphylococcus aureus, Haemophilus influenzae,


and Escherichia coli, are relatively narrow in substrate specificity,
preferring penicillins to cephalosporins. Other β-lactamases, eg,
M AmpC β-lactamase produced by Pseudomonas aeruginosa and
M
L-Ala Enterobacter sp and extended-spectrum β-lactamases (ESBLs) in
Enterobacteriaceae, hydrolyze both cephalosporins and penicil-
L-Ala
R
G
lins. Carbapenems are highly resistant to hydrolysis by peni-
R cillinases and cephalosporinases, but they are hydrolyzed by
G metallo-β-lactamases and carbapenemases.
Altered target PBPs are the basis of methicillin resistance in
M staphylococci and of penicillin resistance in pneumococci and
L-Ala
most resistant enterococci. These resistant organisms produce
M
PBPs that have low affinity for binding β-lactam antibiotics, and
L-Ala +
D-Glu
they are not inhibited except at relatively high, often clinically
G
unachievable, drug concentrations.
G D-Glu L-Lys [Gly]5 Resistance due to impaired penetration of antibiotic occurs
only in Gram-negative species because of the impermeable outer
L-Lys [Gly]5* D-Ala * membrane of their cell wall, which is absent in Gram-positive bac-
R teria. Beta-lactam antibiotics cross the outer membrane and enter
D-Ala D-Ala Gram-negative organisms via outer membrane protein channels
called porins. Absence of the proper channel or down-regulation
D-Ala of its production can greatly impair drug entry into the cell. Poor
Transpeptidase penetration alone is usually not sufficient to confer resistance
because enough antibiotic eventually enters the cell to inhibit
growth. However, this barrier can become important in the pres-
ence of a β-lactamase, even a relatively inefficient one, as long as
M
it can hydrolyze drug faster than it enters the cell. Gram-negative
M L-Ala organisms also may produce an efflux pump, which consists of
R
cytoplasmic and periplasmic protein components that efficiently
L-Ala
G transport some β-lactam antibiotics from the periplasm back
R across the cell wall outer membrane.
G

M Pharmacokinetics
M L-Ala Absorption of orally administered drug differs greatly for indi-
vidual penicillins, depending in part on their acid stability and
L-Ala
G protein binding. Gastrointestinal absorption of nafcillin is erratic,
D-Glu
D-Glu
so it is not suitable for oral administration. Dicloxacillin, ampicil-
G lin, and amoxicillin are acid-stable and relatively well absorbed,
producing serum concentrations in the range of 4–8 mcg/mL after
L-Lys [Gly]5 D-Ala L-Lys [Gly]5
a 500-mg oral dose. Absorption of most oral penicillins (amoxicil-
lin being an exception) is impaired by food, and the drugs should
D-Ala
be administered at least 1–2 hours before or after a meal.
Intravenous administration of penicillin G is preferred to
D-Ala + D-Ala
the intramuscular route because of irritation and local pain
from intramuscular injection of large doses. Serum concen-
FIGURE 43–4  The transpeptidation reaction in Staphylococcus trations 30 minutes after an intravenous injection of 1 g of
aureus that is inhibited by β-lactam antibiotics. The cell wall of Gram- penicillin G (equivalent to approximately 1.6 million units) are
positive bacteria is made up of long peptidoglycan polymer chains 20–50 mcg/mL. Only a fraction of the total drug in serum is
consisting of the alternating aminohexoses N-acetylglucosamine (G) present as free drug, the concentration of which is determined by
and N-acetylmuramic acid (M) with pentapeptide side chains linked (in protein binding. Highly protein-bound penicillins (eg, nafcillin)
S aureus) by pentaglycine bridges. The exact composition of the side
generally achieve lower free-drug concentrations in serum than
chains varies among species. The diagram illustrates small segments of
less protein-bound penicillins (eg, penicillin G or ampicillin).
two such polymer chains and their amino acid side chains. These linear
polymers must be cross-linked by transpeptidation of the side chains at
Penicillins are widely distributed in body fluids and tissues with a
the points indicated by the asterisk to achieve the strength necessary for few exceptions. They are polar molecules, so intracellular concen-
cell viability. trations are well below those found in extracellular fluids.
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     799

Peptidoglycan Amino acid peptide


G = N-acetylglucos-amine
(N-Ag)
G M G M G M G M
Bacterial cell wall M = N-acetylmuramic acid
(N-Am)

Periplasmic space M G M G M G M G

Cytoplasmic membrane
G M G M G M G M
Cytoplasm

Schematic of normal bacterial cell wall peptidoglycan


synthesis transpeptidation reaction.

G M + M G G M G M
Transpeptidase

M G M G

Beta-lactams bind the transpeptidase


at the Penicillin Binding Protein site,
resulting in inhibition of transpeptidation,
thus halting peptidoglycan synthesis. No transpeptidation reaction

Transpeptidase β-lactam
G M + M G G M + M G

FIGURE 43–5  Schematic of a bacterial cell wall and normal synthesis of cell wall peptidoglycan via transpeptidation; M, N-acetylmuramic
acid; Glc, glucose; NAcGlc or G, N-acetylglucosamine. Beta-lactams work by binding the transpeptidase at the penicillin-binding protein site,
resulting in inhibition of transpeptidation, thus halting peptidoglycan synthesis.

Benzathine and procaine penicillins are formulated to delay Penicillin is rapidly excreted by the kidneys; small amounts
absorption, resulting in prolonged blood and tissue concentra- are excreted by other routes. Tubular secretion accounts
tions. A single intramuscular injection of 1.2 million units of for about 90% of renal excretion, and glomerular filtration
benzathine penicillin maintains serum levels above 0.02 mcg/mL accounts for the remainder. The normal half-life of penicillin
for 10 days, sufficient to treat β-hemolytic streptococcal infec- G is approximately 30 minutes but, in renal failure, may be
tions. After 3 weeks, levels still exceed 0.003 mcg/mL, which is as long as 10 hours. Ampicillin and the extended-spectrum
enough to prevent most β-hemolytic streptococcal infections. A penicillins are secreted more slowly than penicillin G and
600,000-unit dose of procaine penicillin yields peak concentra- have half-lives of 1 hour. For penicillins that are cleared by the
tions of 1–2 mcg/mL and clinically useful concentrations for kidney, the dose must be adjusted according to renal function,
12–24 hours after a single intramuscular injection. with approximately one-fourth to one-third the normal dose
Penicillin concentrations in most tissues are equal to those in being administered if creatinine clearance is 10 mL/min or less
serum. Penicillin is also excreted into sputum and breast milk to (Table 43–1).
levels 3–15% of those in the serum. Penetration into the eye, the Nafcillin is primarily cleared by biliary excretion. Oxacillin,
prostate, and the central nervous system is poor. However, with dicloxacillin, and cloxacillin are eliminated by both the kidney
active inflammation of the meninges, as in bacterial meningitis, and biliary excretion, and no dosage adjustment is required for
penicillin concentrations of 1–5 mcg/mL can be achieved with these drugs in patients in renal failure. Because clearance of peni-
a daily parenteral dose of 18–24 million units. These concentra- cillins is less efficient in the newborn, doses adjusted for weight
tions are sufficient to kill susceptible strains of pneumococci and alone result in higher systemic concentrations for longer periods
meningococci. than in the adult.
800    SECTION VIII  Chemotherapeutic Drugs

TABLE 43–1  Guidelines for dosing of some commonly used penicillins.


Adjusted Dose as a Percentage of
Normal Dose for Renal Failure Based
on Creatinine Clearance (Clcr)

Antibiotic (Route of Clcr Approx Clcr Approx


Administration) Adult Dose Pediatric Dose1 Neonatal Dose2 50 mL/min 10 mL/min

Penicillins
  Penicillin G (IV) 1–4 × 106 units 25,000–400,000 units/kg/d 75,000–150,000 units/ 50–75% 25%
q4–6h in 4–6 doses kg/d in 2 or 3 doses
  Penicillin V (PO) 0.25–0.5 g qid 25–75 mg/kg/d in 4 doses   None None
Antistaphylococcal penicillins
 Cloxacillin, 0.25–0.5 g qid 15–25 mg/kg/d in 4 doses   100% 100%
dicloxacillin (PO)
  Nafcillin (IV) 1–2 g q4–6h 100–200 mg/kg/d in 50–75 mg/kg/d in 100% 100%
4–6 doses 2 or 3 doses
  Oxacillin (IV) 1–2 g q4–6h 50–100 mg/kg/d in 50–75 mg/kg/d in 100% 100%
4–6 doses 2 or 3 doses
Extended-spectrum penicillins
  Amoxicillin (PO) 0.25–0.5 g tid 20–40 mg/kg/d in 3 doses   66% 33%
 Amoxicillin/potassium 500/125 mg tid– 20–40 mg/kg/d in 3 doses   66% 33%
clavulanate (PO) 875/125 mg bid
3
 Piperacillin/ 3.375–4.5 g q4–6h 300 mg/kg/d in 4–6 doses 150 mg/kg/d in 50–75% 25–33%
tazobactam (IV) 2 doses3
1
The total dose should not exceed the adult dose.
2
The dose shown is during the first week of life. The daily dose should be increased by approximately 33–50% after the first week of life. The lower dosage range should be used
for neonates weighing less than 2 kg. After the first month of life, pediatric doses may be used.
3
Dose is based on piperacillin component.

Clinical Uses Penicillin V, the oral form of penicillin, is indicated only in


minor infections because of its relatively poor bioavailability, the
Except for amoxicillin, oral penicillins should be given 1–2 hours
need for dosing four times a day, and its narrow antibacterial spec-
before or after a meal; they should not be given with food to mini-
trum. Amoxicillin (see below) is often used instead.
mize binding to food proteins and acid inactivation. Amoxicillin
Benzathine penicillin and procaine penicillin G for intra-
may be given without regard to meals. Blood levels of all penicil-
muscular injection yield low but prolonged drug levels. A single
lins can be raised by simultaneous administration of probenecid,
intramuscular injection of benzathine penicillin, 1.2 million units,
0.5 g (10 mg/kg in children) every 6 hours orally, which impairs
is effective treatment for β-hemolytic streptococcal pharyngitis.
renal tubular secretion of weak acids such as β-lactam compounds.
Given intramuscularly once every 3–4 weeks, it prevents reinfec-
Penicillins, like all antibacterial antibiotics, should never be used
tion. Benzathine penicillin G, 2.4 million units intramuscularly
for viral infections and should be prescribed only when there is
once a week for 1–3 weeks, is effective in the treatment of syphilis.
reasonable suspicion of, or documented infection with, susceptible
Procaine penicillin G was once a commonly used treatment for
organisms.
pneumococcal pneumonia and gonorrhea; however, it is rarely
used now because many gonococcal strains are penicillin-resistant,
A. Penicillin
and many pneumococci require higher doses of penicillin G or the
Penicillin G is a drug of choice for infections caused by strep- use of more potent β-lactams.
tococci, meningococci, some enterococci, penicillin-susceptible
pneumococci, staphylococci confirmed to be non-β-lactamase-
producing, Treponema pallidum and certain other spirochetes, B. Penicillins Resistant to Staphylococcal Beta-
some Clostridium species, Actinomyces and certain other Gram- Lactamase (Methicillin, Nafcillin, and Isoxazolyl Penicillins)
positive rods, and non-β-lactamase-producing Gram-negative These semisynthetic penicillins are indicated for infections caused
anaerobic organisms. Depending on the organism, the site, and by β-lactamase-producing staphylococci, although penicillin-
the severity of infection, effective doses range between 4 and susceptible strains of streptococci and pneumococci are also sus-
24 million units per day administered intravenously in four to ceptible to these agents. Listeria monocytogenes, enterococci, and
six divided doses. High-dose penicillin G can also be given as a methicillin-resistant strains of staphylococci are resistant. In recent
continuous intravenous infusion. years the empirical use of these drugs has decreased substantially
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     801

because of increasing rates of methicillin resistance in staphylo- therapy, an antipseudomonal β-lactam is sometimes used in com-
cocci. However, for infections caused by methicillin-susceptible bination with an aminoglycoside or fluoroquinolone, particularly
and penicillin-resistant strains of staphylococci, these are consid- in infections outside the urinary tract, despite a lack of data sup-
ered drugs of choice. porting combination therapy over single-drug therapy.
An isoxazolyl penicillin such as dicloxacillin, 0.25–0.5 g orally Ampicillin, amoxicillin, piperacillin, and, historically, ticarcil-
every 4–6 hours (15–25 mg/kg/d for children), is suitable for lin, are available in combination with one of several β-lactamase
treatment of mild to moderate localized staphylococcal infections. inhibitors: clavulanic acid, sulbactam, or tazobactam. The
These drugs are relatively acid-stable and have reasonable bioavail- addition of a β-lactamase inhibitor extends the activity of these
ability. However, food interferes with absorption, and the drugs penicillins to include β-lactamase-producing strains of S aureus as
should be administered 1 hour before or after meals. well as some β-lactamase-producing Gram-negative bacteria (see
Methicillin, the first antistaphylococcal penicillin to be devel- Beta-Lactamase Inhibitors).
oped, is no longer used clinically due to high rates of adverse
effects. Oxacillin and nafcillin, 8–12 g/d, given by intermittent Adverse Reactions
intravenous infusion of 1–2 g every 4–6 hours (50–200 mg/kg/d
The penicillins are generally well tolerated, and, unfortunately,
for children), are considered drugs of choice for serious staphylo-
this may encourage inappropriate use. Most of the serious adverse
coccal infections such as endocarditis.
effects are due to hypersensitivity. The antigenic determinants are
degradation products of penicillins, particularly penicilloic acid
C. Extended-Spectrum Penicillins (Aminopenicillins, and products of alkaline hydrolysis bound to host protein. A his-
Carboxypenicillins, and Ureidopenicillins) tory of a penicillin reaction is not reliable. About 5–8% of people
These drugs have greater activity than penicillin against Gram- claim such a history, but only a small number of these will have
negative bacteria because of their enhanced ability to penetrate a serious reaction when given penicillin. Less than 1% of persons
the Gram-negative outer membrane. Like penicillin G, they are who previously received penicillin without incident will have an
inactivated by many β-lactamases. allergic reaction when given penicillin. Because of the potential
The aminopenicillins, ampicillin and amoxicillin, have very for anaphylaxis, however, penicillin should be administered with
similar spectrums of activity, but amoxicillin is better absorbed caution or a substitute drug given if the person has a history of
orally. Amoxicillin, 250–500 mg three times daily, is equivalent to serious penicillin allergy. Penicillin skin testing may also be used
the same amount of ampicillin given four times daily. Amoxicillin to evaluate Type I hypersensitivity. If skin testing is negative, most
is given orally to treat bacterial sinusitis, otitis, and lower respira- patients can safely receive penicillin.
tory tract infections. Ampicillin and amoxicillin are the most active Allergic reactions include anaphylactic shock (very rare—0.05%
of the oral β-lactam antibiotics against pneumococci with elevated of recipients); serum sickness–type reactions (now rare—urticaria,
MICs to penicillin and are the preferred β-lactam antibiotics for fever, joint swelling, angioedema, pruritus, and respiratory com-
treating infections suspected to be caused by these strains. Ampi- promise occurring 7–12 days after exposure); and a variety of skin
cillin (but not amoxicillin) is effective for shigellosis. Ampicillin, rashes. Oral lesions, fever, interstitial nephritis (an autoimmune
at dosages of 4–12 g/d intravenously, is useful for treating serious reaction to a penicillin-protein complex), eosinophilia, hemolytic
infections caused by susceptible organisms, including anaerobes, anemia and other hematologic disturbances, and vasculitis may
enterococci, L monocytogenes, and β-lactamase-negative strains of also occur. Most patients allergic to penicillins can be treated
Gram-negative cocci and bacilli such as E coli, and Salmonella sp. with alternative drugs. However, if necessary (eg, treatment of
Non-β-lactamase-producing strains of H influenzae are generally enterococcal endocarditis or neurosyphilis in a patient with seri-
susceptible, but strains that are resistant because of altered PBPs ous penicillin allergy), desensitization can be accomplished with
are emerging. Due to production of β-lactamases by Gram- gradually increasing doses of penicillin.
negative bacilli, ampicillin can no longer be used for empirical In patients with renal failure, penicillin in high doses can
therapy of urinary tract infections and typhoid fever. Ampicillin cause seizures. Nafcillin is associated with neutropenia and
is not active against Klebsiella sp, Enterobacter sp, P aeruginosa, interstitial nephritis; oxacillin can cause hepatitis; and methi-
Citrobacter sp, Serratia marcescens, indole-positive Proteus species, cillin commonly caused interstitial nephritis (and is no longer
and other Gram-negative aerobes that are commonly encountered used for this reason). Large doses of penicillins given orally may
in hospital-acquired infections. These organisms intrinsically pro- lead to gastrointestinal upset, particularly nausea, vomiting, and
duce β-lactamases that inactivate ampicillin. diarrhea. Ampicillin has been associated with pseudomembra-
The carboxypenicillins, carbenicillin and ticarcillin, were nous colitis. Secondary infections such as vaginal candidiasis
developed to broaden the spectrum of penicillins against Gram- may occur. Ampicillin and amoxicillin can be associated with
negative pathogens, including P aeruginosa; however, neither skin rashes when prescribed in the setting of viral illnesses,
agent is available in the USA. The ureidopenicillin piperacillin is particularly noted during acute Epstein-Barr virus infection,
also active against many Gram-negative bacilli, such as Klebsiella but the incidence of rash may be lower than originally reported.
pneumoniae and P aeruginosa. Piperacillin is available only as a Piperacillin-tazobactam, when combined with vancomycin, has
co-formulation with the β-lactamase inhibitor tazobactam. Due been associated with greater incidence of acute kidney injury
to the propensity of P aeruginosa to develop resistance during compared to alternate β-lactam agents.
802    SECTION VIII  Chemotherapeutic Drugs

■■ CEPHALOSPORINS & O

CEPHAMYCINS R1 C NH
B
S
A
N R2
O
Cephalosporins are similar to penicillins but are more stable COO

to many bacterial β-lactamases and, therefore, have a broader N N


R1 R2
N N
spectrum of activity. However, strains of E coli and Klebsiella sp Cefazolin N CH2
CH2 S CH3
expressing extended-spectrum β-lactamases that can hydrolyze N S

most cephalosporins are a growing clinical concern. Cephalo- Cephalexin CH CH3

sporins are not active against L monocytogenes, and of the avail- NH2

able cephalosporins, only ceftaroline has some activity against Cefadroxil HO CH CH3
enterococci. NH2

O
Cefoxitin
Chemistry S
CH2 CH2 O C NH2

The nucleus of the cephalosporins, 7-aminocephalosporanic acid Cefaclor CH Cl


(Figure 43–6), bears a close resemblance to 6-aminopenicillanic NH2
acid (Figure 43–1). The intrinsic antimicrobial activity of HO

natural cephalosporins is low, but the attachment of various R1 Cefprozil CH CH CH3

and R2 groups has yielded hundreds of potent compounds, many O


NH
C 2
O
with low toxicity. Cephalosporins have traditionally been classi- Cefuroxime O
N CH2 C NH2
fied into four major groups or generations, depending mainly O
OCH3
N N
on the spectrum of antimicrobial activity. Several cephalosporins 1 H2NC C C S C
CH2 S N
Cefotetan N
developed more recently do not fit the traditional classification HOOC S
CH3
groups. Their unique characteristics and spectra of activity are N C
O
Cefotaxime
outlined below. H 2N S
N
OCH3
CH2 O C CH3

N C
1
Cefpodoxime N
CH2 O CH3

FIRST-GENERATION CEPHALOSPORINS H 2N
O
S
OH
OCH3

C
First-generation cephalosporins include cefazolin, cefadroxil, Ceftibuten
C H
N
cephalexin, cephalothin, cephapirin, and cephradine; cefazolin H2N
S
and cephalexin are the only two available in the USA. These drugs H2N
OH
S
are very active against Gram-positive cocci, such as streptococci Cefdinir
N CH CH2
N
and staphylococci. Traditional cephalosporins are not active C
H
N C H3C
against methicillin-resistant strains of staphylococci; however, Ceftriaxone N
N N O

new compounds have been developed that have activity against H2N S OCH3
N O
CH2 S
methicillin-resistant strains (see below). E coli, K pneumoniae, and N C
CH3
N
Proteus mirabilis are often sensitive to first-generation cephalo- Ceftazidime
H2N S O C COOH
CH2 N
sporins, but activity against P aeruginosa, indole-positive Proteus CH3

species, Enterobacter sp, S marcescens, Citrobacter sp, and Acineto- N C


CH2 N+
bacter sp is poor. Anaerobic cocci (eg, peptococci, peptostrepto- Cefepime
H2N S
N
OCH3 CH3
cocci) are usually sensitive, but Bacteroides fragilis is not.
O
N N+ CH3
N N
Pharmacokinetics & Dosage Ceftaroline S
N
S
S
H2N
A. Oral S
H2N N
Cephalexin is the oral first generation agent widely used in N
the USA. After oral doses of 500 mg, peak serum levels are N O
Ceftolozane R1 NH
15–20 mcg/mL. Urine concentration is usually very high, but in O N
NH
most tissues levels are variable and generally lower than in serum. H3C OH
H3C
N
N
H 3C
Cephalexin is typically given in oral dosages of 0.25–0.5 g four O

times daily (15–30 mg/kg/d). Excretion is mainly by glomerular


filtration and tubular secretion into the urine. Drugs that block FIGURE 43–6  Structures of some cephalosporins. R1 and R2
tubular secretion, eg, probenecid, may increase serum levels sub- structures are substituents on the 7-aminocephalosporanic acid
stantially. In patients with impaired renal function, dosage must nucleus pictured at the top. Other structures (cefoxitin and below)
be reduced (Table 43–2). are complete in themselves. 1Additional substituents not shown.
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     803

TABLE 43–2  Guidelines for dosing of some commonly used cephalosporins and other cell-wall inhibitor antibiotics.
Adjusted Dose as a Percentage
of Normal Dose for Renal Failure
Based on Creatinine Clearance (Clcr)

Antibiotic (Route of Clcr Approx Clcr Approx


Administration) Adult Dose Pediatric Dose1 Neonatal Dose2 50 mL/min 10 mL/min

First-generation cephalosporins
  Cephalexin (PO) 0.25–0.5 g qid 25–50 mg/kg/d in 4 doses   50% 25%
  Cefazolin (IV) 0.5–2 g q8h 25–100 mg/kg/d in 3 or 4   50% 25%
doses
Second-generation cephalosporins
  Cefoxitin (IV) 1–2 g q6–8h 75–150 mg/kg/d in 3 or 4   50–75% 25%
doses
  Cefotetan (IV) 1–2 g q12h     50% 25%
  Cefuroxime (IV) 0.75–1.5 g q8h 50–100 mg/kg/d in 3 or   66% 25–33%
4 doses
Third- and fourth-generation cephalosporins including ceftaroline fosamil
  Cefotaxime (IV) 1–2 g q6–12h 50–200 mg/kg/d in 4–6 doses 100 mg/kg/d in 2 doses 50% 25%
  Ceftazidime (IV) 1–2 g q8–12h 75–150 mg/kg/d in 3 doses 100–150 mg/kg/d in 50% 25%
2 or 3 doses
  Ceftriaxone (IV) 1–4 g q24h 50–100 mg/kg/d in 1 or 50 mg/kg/d qd None None
2 doses
  Cefepime (IV) 0.5–2 g q12h 75–120 mg/kg/d in 2 or   50% 25%
3 divided doses
  Ceftaroline fosamil (IV) 600 mg q12h     50–66% 33%
Cephalosporin–β-lactamase inhibitor combinations
 Ceftazidime- 2.5 g q8h     25–50% 6.25–12.5%
avibactam (IV)
 Ceftolozane- 1.5 g q8h     25–50% Not studied
tazobactam (IV)
Carbapenems
  Ertapenem (IM or IV) 1 g q24h     100%3 50%
 Doripenem 500 mg q8h     50% 33%
  Imipenem (IV) 0.25–0.5 g q6–8h     75% 50%
  Meropenem (IV) 1 g q8h (2 g q8h 60–120 mg/kg/d in 3 doses   66% 50%
for meningitis) (maximum of 2 g q8h)
Glycopeptides
  Vancomycin (IV) 30–60 mg/kg/d in 40 mg/kg/d in 3 or 4 doses 15 mg/kg load, then 40% 10%
2–3 doses 20 mg/kg/d in 2 doses
  Telavancin (IV) 10 mg/kg daily     75% 50%
  Dalbavancin (IV) 1000 mg on day 1,     None 75%
500 mg day 8 >30 mL/min
Alternative:
1500 mg × 1
  Oritavancin (IV) 1200 mg × 1     None Not studied
>30 mL/min
Lipopeptides (IV)
 Daptomycin 4–6 mg/kg IV daily     None 50%
>30 mL/min
1
The total dose should not exceed the adult dose.
2
The dose shown is during the first week of life. The daily dose should be increased by approximately 33–50% after the first week of life. The lower dosage range should be used
for neonates weighing less than 2 kg. After the first month of life, pediatric doses may be used.
3
50% of dose for Clcr <30 mL/min.
804    SECTION VIII  Chemotherapeutic Drugs

B. Parenteral Pharmacokinetics & Dosage


Cefazolin is the only first-generation parenteral cephalosporin A. Oral
still in general use. After an intravenous infusion of 1 g, the
Cefuroxime axetil is the most commonly used oral cephalosporin
peak level of cefazolin is approximately 185 mcg/mL. The usual
in the USA. The usual dosage for adults is 250–500 mg orally
intravenous dosage of cefazolin for adults is 0.5–2 g intravenously
twice daily; children should be given 20–40 mg/kg/d up to a
every 8 hours. Cefazolin can also be administered intramuscularly.
maximum of 1 g/d. These drugs are not predictably active against
Excretion is via the kidney, and dose adjustments must be made
penicillin-non-susceptible pneumococci.
for impaired renal function.
B. Parenteral
Clinical Uses After a 1-g intravenous infusion, serum levels are 75–125 mcg/mL
Oral drugs may be used for the treatment of urinary tract infec- for most second-generation cephalosporins. Intramuscular admin-
tions and staphylococcal or streptococcal infections, including cel- istration is painful and should be avoided. Doses and dosing
lulitis or soft tissue abscess. However, oral cephalosporins should intervals vary depending on the specific agent (Table 43–2). There
not be relied on in serious systemic infections. are differences in half-life, protein binding, and interval between
Cefazolin penetrates well into most tissues. It is a drug of choice doses. All are renally cleared and require dosage adjustment in
for surgical prophylaxis and for many streptococcal and staphylo- renal failure.
coccal infections requiring intravenous therapy. Cefazolin may be
used for infections due to E coli or K pneumoniae when the organ- Clinical Uses
ism has been documented to be susceptible. Cefazolin does not
penetrate the central nervous system and cannot be used to treat The oral second-generation cephalosporins are active against
meningitis. Cefazolin is better tolerated than antistaphylococcal β-lactamase-producing H influenzae or Moraxella catarrhalis and
penicillins, and it has been shown to be effective for serious staphy- have been used primarily to treat sinusitis, otitis, and lower respi-
lococcal infections, eg, bacteremia. It can also be used in patients ratory tract infections. Because of their activity against anaerobes
with mild penicillin allergy other than immediate hypersensitivity. (including many B fragilis strains), cefoxitin and cefotetan can be
used to treat mixed anaerobic infections such as peritonitis, diver-
ticulitis, and pelvic inflammatory disease. Cefuroxime is some-
SECOND-GENERATION times used to treat community-acquired pneumonia because it is
active against β-lactamase-producing H influenzae and also many
CEPHALOSPORINS pneumococci. Although cefuroxime crosses the blood-brain bar-
rier, it is less effective in treatment of meningitis than ceftriaxone
Members of the second-generation cephalosporins include
or cefotaxime and should not be used.
cefaclor, cefamandole, cefonicid, cefuroxime, cefprozil,
loracarbef, and ceforanide—of which cefaclor, cefuroxime,
and cefprozil are available in the USA—and the structurally
related cephamycins cefoxitin and cefotetan, which have activity
THIRD-GENERATION CEPHALOSPORINS
against anaerobes. This is a heterogeneous group with individual Third-generation agents include cefoperazone, cefotaxime, ceftazi-
differences in activity, pharmacokinetics, and toxicity. In general, dime, ceftizoxime, ceftriaxone, cefixime, cefpodoxime proxetil,
second-generation cephalosporins are relatively active against cefdinir, cefditoren pivoxil, ceftibuten, and moxalactam. Cefo-
organisms inhibited by first-generation drugs, but, in addition, perazone, ceftizoxime, and moxalactam are no longer commer-
they have extended Gram-negative coverage. Klebsiella sp (includ- cially available in the USA.
ing those resistant to first-generation cephalosporins) are usually
sensitive. Cefuroxime and cefaclor are active against H influenzae
but not against Serratia or B fragilis. In contrast, cefoxitin and Antimicrobial Activity
cefotetan are active against B fragilis and some Serratia strains Compared with second-generation agents, these drugs have
but are less active against H influenzae. As with first-generation expanded Gram-negative coverage, and some are able to cross the
agents, no member of this group is active against enterococci or blood-brain barrier. Third-generation drugs may be active against
P aeruginosa. Compared with other cephalosporins, cefoxitin Citrobacter, S marcescens, and Providencia. They are also effective
shows improved stability in the presence of extended-spectrum against β-lactamase-producing strains of Haemophilus and Neis-
β-lactamases produced by E coli and Klebsiella sp. Clinical data seria. Ceftazidime is the only agent with useful activity against
are limited, but it may offer an alternative to carbapenems in P aeruginosa. Like the second-generation drugs, third-generation
treating certain infections due to these organisms. Second- cephalosporins are hydrolyzed by constitutively produced AmpC
generation cephalosporins may exhibit in vitro activity against β-lactamase, and they are not reliably active against Enterobacter
Enterobacter sp, but resistant mutants that constitutively express a species. Serratia, Providencia, Acinetobacter, and Citrobacter also
chromosomal β-lactamase that hydrolyzes these compounds (and produce a chromosomally encoded cephalosporinase that, when
third-generation cephalosporins) are readily selected, and they constitutively expressed, can confer resistance to third-generation
should not be used to treat Enterobacter infections. cephalosporins. Cefixime, cefdinir, ceftibuten, and cefpodoxime
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     805

proxetil are oral agents possessing similar activity except that cefix- FOURTH-GENERATION
ime and ceftibuten are much less active against pneumococci and
have poor activity against S aureus.
CEPHALOSPORINS
Cefepime is the only available fourth-generation cephalosporin.
Pharmacokinetics & Dosage It is more resistant to hydrolysis by chromosomal β-lactamases
(eg, those produced by Enterobacter). However, like the third-
Intravenous infusion of 1 g of a parenteral cephalosporin produces generation compounds, it is hydrolyzed by extended-spectrum
serum levels of 60–140 mcg/mL. Third-generation cephalosporins β-lactamases. Cefepime has good activity against P aeruginosa,
penetrate body fluids and tissues well and intravenous cephalospo- Enterobacteriaceae, methicillin-susceptible S aureus, and S pneu-
rins achieve levels in the cerebrospinal fluid sufficient to inhibit moniae. It is highly active against Haemophilus and Neisseria sp. It
most susceptible pathogens. penetrates well into cerebrospinal fluid. It is cleared by the kidneys
The half-lives of these drugs and the necessary dosing and has a half-life of 2 hours, and its pharmacokinetic proper-
intervals vary greatly: ceftriaxone (half-life 7–8 hours) can be ties are very similar to those of ceftazidime. Unlike ceftazidime,
injected once every 24 hours at a dosage of 15–50 mg/kg/d. A however, cefepime has good activity against most penicillin-non-
single daily 1-g dose is sufficient for most serious infections, susceptible strains of streptococci, and it is useful in treatment of
with 2 g every 12 hours recommended for treatment of men- Enterobacter infections. The standard dose for cefepime is 1–2 g
ingitis and 2 g every 24 hours recommended for endocarditis. infused every 12 hours; however, when treating more complicated
The remaining drugs in the group (half-life 1–1.7 hours) can infections due to P aeruginosa or in the setting of immunocom-
be infused every 6–8 hours in dosages between 2 and 12 g/d, promise, doses are typically increased to 2 g every 8 hours. Because
depending on the severity of infection. Cefixime can be given of its broad-spectrum activity, cefepime is commonly used empiri-
orally (200 mg twice daily or 400 mg once daily) for urinary cally in patients presenting with febrile neutropenia, in combina-
tract infections. Due to increasing resistance, cefixime is no tion with other agents.
longer recommended for the treatment of uncomplicated
gonococcal urethritis and cervicitis. Intramuscular ceftriaxone
in combination with azithromycin is the regimen of choice Cephalosporins Active against Methicillin-
for treating most gonococcal infections. The adult dose for Resistant Staphylococci
cefpodoxime proxetil or cefditoren pivoxil is 200–400 mg Beta-lactam antibiotics with activity against methicillin-resistant
twice daily; for ceftibuten, 400 mg once daily; and for cefdinir, staphylococci are currently under development. Ceftaroline
300 mg/12 h. Ceftriaxone excretion is mainly through the fosamil, the prodrug of the active metabolite ceftaroline, is the
biliary tract, and no dosage adjustment is required in renal first such drug to be approved for clinical use in the USA. Cef-
insufficiency. The other third-generation cephalosporins are taroline has increased binding to penicillin-binding protein 2a,
excreted by the kidney and therefore require dosage adjustment which mediates methicillin resistance in staphylococci, resulting
in renal insufficiency. in bactericidal activity against these strains. It has some in vitro
activity against enterococci and a broad Gram-negative spectrum
similar to ceftriaxone. It is not active against AmpC or extended-
Clinical Uses spectrum β-lactamase-producing organisms. Ceftaroline is cur-
Third-generation cephalosporins are used to treat a wide variety of rently approved for the treatment of skin and soft tissue infections
serious infections caused by organisms that are resistant to most and community-acquired pneumonia at a dose of 600 mg infused
other drugs. Strains expressing extended-spectrum β-lactamases, every 12 hours. It has been used off-label to treat complicated
however, are not susceptible. Third-generation cephalosporins infections such as bacteremia, endocarditis, and osteomyelitis,
should be avoided in treatment of Enterobacter infections—even sometimes in combination with other agents and often at an
if the clinical isolate appears susceptible in vitro—because of increased dose of 600 mg every 8 hours. The normal half-life
emergence of resistance. Ceftriaxone and cefotaxime are approved is about 2.7 hours; ceftaroline is primarily excreted renally and
for treatment of meningitis, including meningitis caused by pneu- requires dose adjustment in renal impairment.
mococci, meningococci, H influenzae, and susceptible enteric
Gram-negative rods, but not by L monocytogenes. Ceftriaxone and
Cephalosporins Combined with
cefotaxime are the most active cephalosporins against penicillin-
non-susceptible strains of pneumococci and are recommended a-lactamase Inhibitors
for empirical therapy of serious infections that may be caused Novel cephalosporin-β-lactamase inhibitor combinations have
by these strains. Meningitis caused by strains of pneumococci been developed to combat resistant Gram-negative infections;
with penicillin MICs >1 mcg/mL may not respond even to these see the subsequent section for more information on β-lactamase
agents, and addition of vancomycin is recommended. Other inhibitors. Ceftolozane-tazobactam and ceftazidime-avibactam
potential indications include empirical therapy of sepsis in both were both FDA-approved for the treatment of complicated
the immunocompetent and the immunocompromised patient intra-abdominal infections and urinary tract infections. Both
and treatment of infections for which a cephalosporin is the least agents have potent in vitro activity against Gram-negative organ-
toxic drug available. isms, including P aeruginosa and AmpC and extended-spectrum
806    SECTION VIII  Chemotherapeutic Drugs

β-lactamase producing Enterobacteriaceae. While neither agent is reactions; consequently, alcohol and alcohol-containing medica-
active against organisms producing metallo-β-lactamases, ceftazi- tions must be avoided.
dime-avibactam may be an option for carbapenemase-producing
organisms. Due to limited activity against anaerobic pathogens,
both should be combined with metronidazole when treating ■■ OTHER BETA-LACTAM DRUGS
complicated intra-abdominal infections. Both agents have short
half-lives of 2–3 hours and are dosed every 8 hours. Both are MONOBACTAMS
primarily renally excreted and require dose adjustment in patients
with impaired renal clearance. Monobactams are drugs with a monocyclic β-lactam ring
(Figure 43–1). Their spectrum of activity is limited to aerobic
Gram-negative organisms (including P aeruginosa). Unlike other
ADVERSE EFFECTS OF β-lactam antibiotics, they have no activity against Gram-positive
CEPHALOSPORINS bacteria or anaerobes. Aztreonam is the only monobactam avail-
able in the USA. It has structural similarities to ceftazidime,
A. Allergy and its Gram-negative spectrum is similar to that of the third-
Like penicillins, cephalosporins may elicit a variety of hyper- generation cephalosporins. It is stable to many β-lactamases with
sensitivity reactions, including anaphylaxis, fever, skin rashes, notable exceptions being AmpC β-lactamases and extended-
nephritis, granulocytopenia, and hemolytic anemia. Patients spectrum β-lactamases. It penetrates well into the cerebrospinal
with documented penicillin anaphylaxis have an increased risk fluid. Aztreonam is given intravenously every 8 hours in a dose of
of reacting to cephalosporins compared with patients without a 1–2 g, providing peak serum levels of 100 mcg/mL. The half-life
history of penicillin allergy. However, the chemical nucleus of is 1–2 hours and is greatly prolonged in renal failure.
cephalosporins is sufficiently different from that of penicillins Penicillin-allergic patients tolerate aztreonam without reaction.
such that many individuals with a history of penicillin allergy tol- Notably, because of its structural similarity to ceftazidime, there is
erate cephalosporins. Overall, the frequency of cross-allergenicity potential for cross-reactivity; aztreonam should be used with caution
between the two groups of drugs is low (~1%). Cross-allergenicity in the case of documented severe allergies to ceftazidime. Occasional
appears to be most common among penicillin, aminopenicillins, skin rashes and elevations of serum aminotransferases occur during
and early-generation cephalosporins, which share similar R-1 side administration of aztreonam, but major toxicity is uncommon. In
chains. Patients with a history of anaphylaxis to penicillins should patients with a history of penicillin anaphylaxis, aztreonam may be
not receive first- or second-generation cephalosporins, while third- used to treat serious infections such as pneumonia, meningitis, and
and fourth-generation cephalosporins should be administered sepsis caused by susceptible Gram-negative pathogens.
with caution, preferably in a monitored setting.

B. Toxicity BETA-LACTAMASE INHIBITORS


Local irritation can produce pain after intramuscular injection (CLAVULANIC ACID, SULBACTAM,
and thrombophlebitis after intravenous injection. Renal toxicity, TAZOBACTAM, & AVIBACTAM)
including interstitial nephritis and tubular necrosis, may occur
uncommonly. Traditional β-lactamase inhibitors (clavulanic acid, sulbactam,
Cephalosporins that contain a methylthiotetrazole group may and tazobactam) resemble β-lactam molecules (Figure 43–7), but
cause hypoprothrombinemia and bleeding disorders. Historically, they have very weak antibacterial action. They are potent inhibi-
this group included cefamandole, cefmetazole, and cefoperazone; tors of many but not all bacterial β-lactamases and can protect
however, cefotetan is the only methylthiotetrazole-containing hydrolyzable penicillins from inactivation by these enzymes. The
agent used in the USA. Oral administration of vitamin K, 10 mg traditional β-lactamase inhibitors are most active against Ambler
twice weekly, can prevent this uncommon problem. Drugs with class A β-lactamases (plasmid-encoded transposable element
the methylthiotetrazole ring can also cause severe disulfiram-like [TEM] β-lactamases in particular), such as those produced by

– O
O O
S C
CH3
H2C CH O H2C CH H 2N
CH2 R
C N C N
C N
O CH CH2OH O COOH R= N N C N
COOH R=H N O OSO3–
Clavulanic acid Sulbactam Tazobactam Avibactam

FIGURE 43–7  Beta-lactamase inhibitors.


CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     807

staphylococci, H influenzae, N gonorrhoeae, Salmonella, Shigella, dosage of doripenem is 0.5 g administered as a 1- or 4-hour infu-
E coli, and K pneumoniae. They are not good inhibitors of class sion every 8 hours. Ertapenem has the longest half-life (4 hours)
C β-lactamases, which typically are chromosomally encoded and is administered as a once-daily dose of 1 g intravenously or
and inducible, produced by Enterobacter sp, Citrobacter sp, intramuscularly. Intramuscular ertapenem is irritating, and the drug
S marcescens, and P aeruginosa, but they do inhibit chromosomal is formulated with 1% lidocaine for administration by this route.
β-lactamases of B fragilis and M catarrhalis. The novel non-β- A carbapenem is indicated for infections caused by suscep-
lactam β-lactamase inhibitor avibactam is active against Ambler tible organisms that are resistant to other available drugs, eg,
class A β-lactamases but also active against Ambler class C and P aeruginosa, and for treatment of mixed aerobic and anaerobic
some Ambler class D β-lactamases. infections. Carbapenems are active against many penicillin-non-
Beta-lactamase inhibitors are available only in fixed combi- susceptible strains of pneumococci. Carbapenems are highly
nations with specific penicillins and cephalosporins. (The fixed active in the treatment of Enterobacter infections because they
combinations available in the USA are listed in Preparations are resistant to destruction by the β-lactamase produced by these
Available.) An inhibitor extends the spectrum of its compan- organisms. Clinical experience suggests that carbapenems are also
ion β-lactam provided that the inactivity against a particular the treatment of choice for serious infections caused by extended-
organism is due to destruction by a β-lactamase and that the spectrum β-lactamase-producing Gram-negative bacteria. Ertape-
inhibitor is active against the β-lactamase that is produced. Thus, nem is insufficiently active against P aeruginosa and should not
ampicillin-sulbactam is active against β-lactamase-producing be used to treat infections caused by this organism. Imipenem,
S aureus and H influenzae but not against Serratia, which produces meropenem, or doripenem, with or without an aminoglycoside,
a β-lactamase that is not inhibited by sulbactam. Similarly, if a may be effective treatment for febrile neutropenic patients.
strain of P aeruginosa is resistant to piperacillin, it is also resistant The most common adverse effects of carbapenems—which
to piperacillin-tazobactam because tazobactam does not inhibit tend to be more common with imipenem—are nausea, vomiting,
the chromosomal β-lactamase produced by P aeruginosa. diarrhea, skin rashes, and reactions at the infusion sites. Exces-
Beta-lactam–β-lactamase inhibitor combinations are fre- sive levels of imipenem in patients with renal failure may lead
quently used as empirical therapy for infections caused by a wide to seizures. Meropenem, doripenem, and ertapenem are much
range of potential pathogens in both immunocompromised and less likely to cause seizures than imipenem. Patients allergic to
immunocompetent patients. Adjustments for renal insufficiency penicillins may be allergic to carbapenems, but the incidence of
are made based on the β-lactam component. cross-reactivity is thought to be less than 1%.

CARBAPENEMS ■■ GLYCOPEPTIDE ANTIBIOTICS


The carbapenems are structurally related to other β-lactam anti- VANCOMYCIN
biotics (Figure 43–1). Doripenem, ertapenem, imipenem, and
meropenem are licensed for use in the USA. Imipenem, the first Vancomycin is an antibiotic isolated from the bacterium now
drug of this class, has a wide spectrum with good activity against known as Amycolatopsis orientalis. It is active primarily against
most Gram-negative rods, including P aeruginosa, Gram-positive Gram-positive bacteria due to its large molecular weight and
organisms, and anaerobes. It is resistant to most β-lactamases but lack of penetration through Gram-negative cell membranes. The
not carbapenemases or metallo-β-lactamases. Enterococcus faecium, intravenous product is water soluble and stable for 14 days in the
methicillin-resistant strains of staphylococci, Clostridium difficile, refrigerator following reconstitution.
Burkholderia cepacia, and Stenotrophomonas maltophilia are resistant.
Imipenem is inactivated by dehydropeptidases in renal tubules, Mechanisms of Action & Basis of
resulting in low urinary concentrations. Consequently, it is admin-
istered together with an inhibitor of renal dehydropeptidase, cilas- Resistance
tatin, for clinical use. Doripenem and meropenem are similar to Vancomycin inhibits cell wall synthesis by binding firmly to the
imipenem but have slightly greater activity against Gram-negative d-Ala-d-Ala terminus of nascent peptidoglycan pentapeptide
aerobes and slightly less activity against Gram-positives. They are (Figure 43–8). This inhibits the transglycosylase, preventing
not significantly degraded by renal dehydropeptidase and do not further elongation of peptidoglycan and cross-linking. The pep-
require an inhibitor. Unlike the other carbapenems, ertapenem does tidoglycan is thus weakened, and the cell becomes susceptible to
not have appreciable activity against P aeruginosa and Acinetobacter lysis. The cell membrane is also damaged, which contributes to
species. It is not degraded by renal dehydropeptidase. the antibacterial effect.
Carbapenems penetrate body tissues and fluids well, including Resistance to vancomycin in enterococci is due to modifica-
the cerebrospinal fluid for all but ertapenem. All are cleared renally, tion of the d-Ala-d-Ala binding site of the peptidoglycan building
and the dose must be reduced in patients with renal insufficiency. block in which the terminal d-Ala is replaced by d-lactate. This
The usual dosage of imipenem is 0.25–0.5 g given intravenously results in the loss of a critical hydrogen bond that facilitates high-
every 6–8 hours (half-life 1 hour). The usual adult dosage of affinity binding of vancomycin to its target and loss of activity.
meropenem is 0.5–1 g intravenously every 8 hours. The usual adult This mechanism is also present in vancomycin-resistant S aureus
808    SECTION VIII  Chemotherapeutic Drugs

Peptidoglycan Amino acid peptide


G = N-acetylglucos-amine
(N-Ag)
G M G M G M G M
Bacterial cell wall M = N-acetylmuramic acid
(N-Am)

Periplasmic space M G M G M G M G

Cytoplasmic membrane
G M G M G M G M
Cytoplasm

Schematic of normal bacterial cell wall peptidoglycan


synthesis transpeptidation reaction.

G M + M G G M G M
Transpeptidase
Crosslinking

M G M G

Vancomycin binds the D-Alanine D-Alanine


terminus of the amino acid peptide, inhibiting V
crosslinkage. V A
A N
N

Vancomycin
G M + M G G M M G

V No crosslinking
A
N

FIGURE 43–8  Schematic of a bacterial cell wall and normal synthesis of cell wall peptidoglycan via transpeptidation; M, N-acetylmuramic
acid; Glc, glucose; NAcGlc or G, N-acetylglucosamine. Vancomycin binds the d-Alanine d-Alanine (d-Ala d-Ala) terminus of the amino acid
peptide, inhibiting cross-linkage of the cell wall.

strains (MIC ≥ 16 mcg/mL), which have acquired the enterococcal dividing; the rate is less than that of the penicillins both in vitro
resistance determinants. The underlying mechanism for reduced and in vivo. Vancomycin is synergistic in vitro with gentamicin
vancomycin susceptibility in vancomycin-intermediate strains and streptomycin against Enterococcus faecium and Enterococcus
(MIC = 4–8 mcg/mL) of S aureus is not fully known. However, faecalis strains that do not exhibit high levels of aminoglycoside
these strains have altered cell wall metabolism that results in a resistance. Vancomycin is active against many Gram-positive
thickened cell wall with increased numbers of d-Ala-d-Ala resi- anaerobes including C difficile.
dues, which serve as dead-end binding sites for vancomycin. Van-
comycin is sequestered within the cell wall by these false targets Pharmacokinetics
and may be unable to reach its site of action.
Vancomycin is poorly absorbed from the intestinal tract and is
administered orally only for the treatment of colitis caused by
Antibacterial Activity C difficile. Parenteral doses must be administered intravenously.
Vancomycin is bactericidal for Gram-positive bacteria in con- A 1-hour intravenous infusion of 1 g produces blood levels of
centrations of 0.5–10 mcg/mL. Most pathogenic staphylococci, 15–30 mcg/mL for 1–2 hours. The drug is widely distributed
including those producing β-lactamase and those resistant to naf- in the body including adipose tissue. Cerebrospinal fluid levels
cillin and methicillin, are killed by 2 mcg/mL or less. Vancomycin 7–30% of simultaneous serum concentrations are achieved if there
kills staphylococci relatively slowly and only if cells are actively is meningeal inflammation. Ninety percent of the drug is excreted
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     809

by glomerular filtration. In the presence of renal insufficiency, Administration with another ototoxic or nephrotoxic drug, such
striking accumulation may occur (Table 43–2). In functionally as an aminoglycoside, increases the risk of these toxicities. Ototox-
anephric patients, the half-life of vancomycin is 6–10 days. A icity can be minimized by maintaining peak serum concentrations
significant amount of vancomycin is removed during a standard below 60 mcg/mL. Among the more common reactions is the
hemodialysis run using a high-flux membrane. so-called “red man” syndrome. This infusion-related flushing is
caused by release of histamine. It can be largely prevented by pro-
Clinical Uses longing the infusion period to 1–2 hours (preferred) or pretreat-
ment with an antihistamine such as diphenhydramine.
Important indications for parenteral vancomycin are bloodstream
infections and endocarditis caused by methicillin-resistant staphy-
lococci. However, vancomycin is not as effective as an antistaphy- TEICOPLANIN
lococcal penicillin for treatment of serious infections such as
endocarditis caused by methicillin-susceptible strains. Vancomy- Teicoplanin is a glycopeptide antibiotic that is very similar to
cin in combination with gentamicin is an alternative regimen for vancomycin in mechanism of action and antibacterial spectrum.
treatment of enterococcal endocarditis in a patient with serious Unlike vancomycin, it can be given intramuscularly as well as
penicillin allergy. Vancomycin (in combination with cefotaxime, intravenously. Teicoplanin has a long half-life (45–70 hours),
ceftriaxone, or rifampin) is also recommended for treatment permitting once-daily dosing. This drug is available in Europe but
of meningitis suspected or known to be caused by a penicillin- has not been approved for use in the USA.
resistant strain of pneumococcus. The recommended dosage in
a patient with normal renal function is 30–60 mg/kg/d in two
or three divided doses. The traditional dosing regimen in adults TELAVANCIN
with normal renal function is 1 g every 12 hours (~30 mg/kg/d);
however, this dose will not typically achieve the trough concentra- Telavancin is a semisynthetic lipoglycopeptide derived from van-
tions (15–20 mcg/mL) recommended for serious infections. For comycin. Telavancin is active versus Gram-positive bacteria and
serious infections (see below), a starting dose of 45–60 mg/kg/d has in vitro activity against many strains with reduced susceptibil-
should be given with titration of the dose to achieve trough levels ity to vancomycin. Telavancin has two mechanisms of action. Like
of 15–20 mcg/mL. The dosage in children is 40 mg/kg/d in vancomycin, telavancin inhibits cell wall synthesis by binding to
three or four divided doses. Clearance of vancomycin is directly the d-Ala-d-Ala terminus of peptidoglycan in the growing cell
proportional to creatinine clearance, and the dosage is reduced wall. In addition, it disrupts the bacterial cell membrane potential
accordingly in patients with renal insufficiency. For patients and increases membrane permeability. The half-life of telavancin
receiving hemodialysis, a common dosing regimen is a 1-g load- is approximately 8 hours, which supports once-daily intravenous
ing dose followed by 500 mg after each dialysis session. Patients dosing. The drug is approved for treatment of complicated skin
receiving a prolonged course of therapy should have serum trough and soft tissue infections and hospital-acquired pneumonia at a
concentrations checked. For S aureus infections, recommended dose of 10 mg/kg IV daily. Unlike vancomycin therapy, monitor-
trough concentrations are 10–15 mcg/mL for mild to moderate ing of serum telavancin levels is not required. Telavancin was asso-
infections and 15–20 mcg/mL for more serious infections such as ciated with substantial nephrotoxicity and concern for increased
endocarditis, meningitis, and necrotizing pneumonia. mortality associated with renal impairment in clinical trials, lead-
Oral vancomycin, 0.125–0.5 g every 6 hours, is used to treat ing to boxed warnings. It is potentially teratogenic, so administra-
colitis caused by C difficile. Because of the emergence of vancomy- tion to pregnant women must be avoided.
cin-resistant enterococci and the potential selective pressure of oral
vancomycin for these resistant organisms, metronidazole had been
preferred as initial therapy. However, use of oral vancomycin does
DALBAVANCIN AND ORITAVANCIN
not appear to be a significant risk factor for acquisition of van- Dalbavancin and oritavancin are semisynthetic lipoglycopeptides
comycin-resistant enterococci. Additionally, recent clinical data derived from teicoplanin. Dalbavancin and oritavancin inhibit cell
suggest that vancomycin is associated with higher initial response wall synthesis via the same mechanism of action as vancomycin
rates than metronidazole, particularly for moderate to severe cases and teicoplanin; oritavancin works by additional mechanisms,
of C difficile colitis. Therefore, oral vancomycin may be used as a including disruption of cell membrane permeability and inhibi-
first-line treatment, especially for severe cases. tion of RNA synthesis. Compared with vancomycin, both agents
have lower MICs against many Gram-positive bacteria including
Adverse Reactions methicillin-resistant and vancomycin-intermediate S aureus. Dal-
Adverse reactions with parenteral administration of vancomycin bavancin is not active against most strains of vancomycin-resistant
are encountered fairly frequently. Most reactions are relatively enterococci (VRE). Oritavancin has in vitro activity against VRE,
minor and reversible. Vancomycin is irritating to tissue, resulting but its clinical utility in treating VRE infections remains unclear.
in phlebitis at the site of injection. Chills and fever may occur. Both agents have extremely long half-lives of greater than 10 days,
Ototoxicity is rare but nephrotoxicity is still encountered regu- which allows for once-weekly intravenous administration. Dal-
larly with current preparations, especially with high trough levels. bavancin and oritavancin have been approved for the treatment
810    SECTION VIII  Chemotherapeutic Drugs

of skin and soft tissue infections. There are limited clinical data evidence supporting increased efficacy is lacking. In clinical tri-
supporting the use of dalbavancin for uncomplicated catheter- als, daptomycin was noninferior in efficacy to vancomycin. It
associated bloodstream infections, though it is not approved for can cause myopathy, and creatine phosphokinase levels should be
use in this setting. Dalbavancin was originally approved as a two- monitored weekly. Pulmonary surfactant antagonizes daptomycin,
dose, once-weekly intravenous regimen (1000 mg infused on day and it should not be used to treat pneumonia. Daptomycin can
1 and 500 mg infused on day 8), but a subsequent phase 3 study also cause an allergic pneumonitis in patients receiving prolonged
comparing the two-dose regimen with a single, 1500-mg intra- therapy (>2 weeks). Treatment failures have been reported in
venous dose showed that the single-dose regimen is noninferior. association with an increase in daptomycin MIC during therapy.
The results of this study allowed for updated labelling, making Daptomycin is an effective alternative to vancomycin, and its role
both dalbavancin and oritavancin appropriate for single-dose continues to unfold.
treatments for complicated skin and soft tissue infections. A prac-
tical difference between the two is the infusion time: dalbavancin
can be administered over 30 minutes, while oritavancin must be FOSFOMYCIN
infused over 3 hours. Neither requires dose adjustment in mild
to moderate renal or hepatic impairment, and neither is removed Fosfomycin trometamol, a stable salt of fosfomycin (phosphono-
by dialysis. mycin), inhibits a very early stage of bacterial cell wall synthesis.
An analog of phosphoenolpyruvate, it is structurally unrelated to
any other antimicrobial agent. It inhibits the cytoplasmic enzyme
enolpyruvate transferase by covalently binding to the cysteine resi-
■■ OTHER CELL WALL- OR due of the active site and blocking the addition of phosphoenol-
MEMBRANE-ACTIVE AGENTS pyruvate to UDP-N-acetylglucosamine. This reaction is the first
step in the formation of UDP-N-acetylmuramic acid, the precur-
DAPTOMYCIN sor of N-acetylmuramic acid, which is found only in bacterial cell
walls. The drug is transported into the bacterial cell by glycero-
Daptomycin is a novel cyclic lipopeptide fermentation product of phosphate or glucose 6-phosphate transport systems. Resistance is
Streptomyces roseosporus (Figure 43–9). Its spectrum of activity is due to inadequate transport of drug into the cell.
similar to that of vancomycin except that it may be active against Fosfomycin is active against both Gram-positive and Gram-
vancomycin-resistant strains of enterococci and S aureus. In vitro, negative organisms at concentrations ≥ 125 mcg/mL. Susceptibil-
it has more rapid bactericidal activity than vancomycin. The pre- ity tests should be performed in growth medium supplemented
cise mechanism of action is not fully understood, but it is known with glucose 6-phosphate to minimize false-positive indications of
to bind to the cell membrane via calcium-dependent insertion of resistance. In vitro synergism occurs when fosfomycin is combined
its lipid tail. This results in depolarization of the cell membrane with β-lactam antibiotics, aminoglycosides, or fluoroquinolones.
with potassium efflux and rapid cell death (Figure 43–10). Dap- Fosfomycin trometamol is available in both oral and par-
tomycin is cleared renally. The approved doses are 4 mg/kg/dose enteral formulations, although only the oral preparation is
for treatment of skin and soft tissue infections and 6 mg/kg/dose approved for use in the USA. Oral bioavailability is approxi-
for treatment of bacteremia and endocarditis once daily in patients mately 40%. Peak serum concentrations are 10 mcg/mL and
with normal renal function and every other day in patients with 30 mcg/mL following a 2-g or 4-g oral dose, respectively. The
creatinine clearance of less than 30 mL/min. For serious infec- half-life is approximately 4 hours. The active drug is excreted
tions, many experts recommend using 8–10 mg/kg/dose. These by the kidney, with urinary concentrations exceeding MICs for
higher doses appear to be safe and well tolerated, although most urinary tract pathogens.

L-Asp D-Ala L-Asp Gly D-Ser 3-MeGlu (L-theo)


O
=

L-Orn Gly L-Thr O C L-Kyn

L-Asp L-Asn L-Trp NH

O
=

Decanoic acid

FIGURE 43–9  Structure of daptomycin. (Kyn, deaminated tryptophan.)


CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     811

Daptomycin

Ca2+
Step 1

Ca2+ Step 2 Step 3


Ca2+

K+

FIGURE 43–10  Proposed mechanism of action of daptomycin. Daptomycin first binds to the cytoplasmic membrane (step 1) and then
forms complexes in a calcium-dependent manner (steps 2 and 3). Complex formation causes a rapid loss of cellular potassium, possibly by pore
formation, and membrane depolarization. This is followed by arrest of DNA, RNA, and protein synthesis resulting in cell death. Cell lysis does not
occur.

Fosfomycin is approved for use as a single 3-g dose for treat- skin or on mucous membranes. Bacitracin is commonly associated
ment of uncomplicated lower urinary tract infections (UTI) in with hypersensitivity and should not be applied to wounds for the
women. Limited data in case reports have suggested efficacy in purpose of preventing infection.
males with UTI and prostatitis; in these cases, a 3-g dose has
been given every 3 days for 9 days when treating UTI or 21 days
for prostatitis. There are no supportive data for using fosfomycin CYCLOSERINE
to treat pyelonephritis. The drug appears to be safe for use in
pregnancy. Cycloserine is an antibiotic produced by Streptomyces orchidaceous.
It is water soluble and very unstable at acid pH. Cycloserine
inhibits many Gram-positive and Gram-negative organisms, but it
BACITRACIN is used almost exclusively to treat tuberculosis caused by strains of
Mycobacterium tuberculosis resistant to first-line agents. Cycloser-
Bacitracin is a cyclic peptide mixture first obtained from the Tracy ine is a structural analog of d-alanine and inhibits the incorpora-
strain of Bacillus subtilis in 1943. It is active against Gram-positive tion of d-alanine into peptidoglycan pentapeptide by inhibiting
microorganisms. Bacitracin inhibits cell wall formation by inter- alanine racemase, which converts l-alanine to d-alanine, and
fering with dephosphorylation in cycling of the lipid carrier that d-alanyl-d-alanine ligase. After ingestion of 0.25 g of cycloserine
transfers peptidoglycan subunits to the growing cell wall. There blood levels reach 20–30 mcg/mL—sufficient to inhibit many
is no cross-resistance between bacitracin and other antimicrobial strains of mycobacteria and Gram-negative bacteria. The drug is
drugs. widely distributed in tissues. Most of the drug is excreted in active
Bacitracin is highly nephrotoxic when administered systemi- form into the urine. The dosage for treating tuberculosis is 0.5 to
cally and is only used topically (Chapter 61). Bacitracin is poorly 1 g/d in two or three divided doses.
absorbed, and topical application results in local antibacterial Cycloserine causes serious, dose-related central nervous system
activity. Bacitracin, 500 units/g in an ointment base (often com- toxicity with headaches, tremors, acute psychosis, and convul-
bined with polymyxin or neomycin), is used for the treatment of sions. If oral dosages are maintained below 0.75 g/d, such effects
infections due to mixed bacterial flora in surface lesions of the can usually be avoided.
812    SECTION VIII  Chemotherapeutic Drugs

SUMMARY  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics


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

PENICILLINS
  •  Penicillin G Prevents bacterial cell Rapid bactericidal activity Streptococcal infections, IV administration • rapid renal clearance (half-life
wall synthesis by binding against susceptible bacteria meningococcal infections, 30 min, so requires dosing every 4 h) • Toxicity:
to and inhibiting cell wall neurosyphilis Immediate hypersensitivity, rash, seizures
transpeptidases

  •  Penicillin V: Oral, low systemic levels limit widespread use


  •  Benzathine penicillin, procaine penicillin: Intramuscular, long-acting formulations
  •  Nafcillin, oxacillin: Intravenous, added stability to staphylococcal β-lactamase, biliary clearance
  • Ampicillin, amoxicillin, piperacillin: Greater activity versus Gram-negative bacteria; addition of β-lactamase inhibitor restores activity against many β-lactamase-producing bacteria

CEPHALOSPORINS
  •  Cefazolin Prevents bacterial cell Rapid bactericidal activity Skin and soft tissue IV administration • renal clearance (half-life 1.5 h)
wall synthesis by binding against susceptible bacteria infections, urinary tract • given every 8 h • poor penetration into the
to and inhibiting cell wall infections, surgical central nervous system (CNS) • Toxicity: Rash,
transpeptidases prophylaxis drug fever

  •  Cephalexin: Oral, first-generation drug used for treating skin and soft tissue infections and urinary tract infections
  •  Cefuroxime: Oral and intravenous, second-generation drug, improved activity versus pneumococcus and Haemophilus influenzae
  •  Cefotetan, cefoxitin: Intravenous, second-generation drugs, activity versus Bacteroides fragilis allows for use in abdominal/pelvic infections
  • Ceftriaxone: Intravenous, third-generation drug, mixed clearance with long half-life (6 hours), good CNS penetration, many uses including pneumonia, meningitis,
pyelonephritis, and gonorrhea
  •  Cefotaxime: Intravenous, third-generation, similar to ceftriaxone; however, clearance is renal and half-life is 1 hour
  •  Ceftazidime: Intravenous, third-generation drug, poor Gram-positive activity, good activity versus Pseudomonas aeruginosa
  •  Cefepime: Intravenous, fourth-generation drug, broad activity with improved stability to chromosomal β-lactamases
  •  Ceftaroline: Intravenous, active against methicillin-resistant staphylococci, broad Gram-negative activity not including Pseudomonas aeruginosa
  • Ceftazidime-avibactam, ceftolozane-tazobactam: Intravenous, cephalosporin-β-lactamase inhibitor combination drugs, broad activity with improved stability to
chromosomal β-lactamase and some extended-spectrum β-lactamases

CARBAPENEMS
  • Imipenem- Prevents bacterial cell Rapid bactericidal activity Serious infections such as IV administration • renal clearance (half-life 1 h),
cilastatin wall synthesis by binding against susceptible bacteria pneumonia and sepsis dosed every 6–8 h, cilastatin added to prevent
to and inhibiting cell wall hydrolysis by renal dehydropeptidase • Toxicity:
transpeptidases Seizures especially in renal failure or with high
doses (>2 g/d)

  •  Meropenem, doripenem: Intravenous, similar activity to imipenem; stable to renal dehydropeptidase, lower incidence of seizures
  •  Ertapenem: Intravenous, longer half-life allows for once-daily dosing, lacks activity versus Pseudomonas aeruginosa and Acinetobacter

MONOBACTAMS
  •  Aztreonam Prevents bacterial cell Rapid bactericidal activity Infections caused by aerobic, IV administration • renal clearance half-life 1.5 h
wall synthesis by binding against susceptible bacteria Gram-negative bacteria in • dosed every 8 h • Toxicity: No cross-allergenicity
to and inhibiting cell wall patients with immediate with penicillins
transpeptidases hypersensitivity to penicillins

GLYCOPEPTIDE
  •  Vancomycin Inhibits cell wall synthesis Bactericidal activity against Infections caused by Oral, IV administration • renal clearance (half-life
by binding to the d-Ala-d- susceptible bacteria, slower Gram-positive bacteria 6 h) • starting dose of 30 mg/kg/d in two or three
Ala terminus of nascent kill than β-lactam including sepsis, endocarditis, divided doses in patients with normal renal
peptidoglycan antibiotics and meningitis • C difficile function • trough concentrations of
colitis (oral formulation) 10–15 mcg/mL sufficient for most infections
• Toxicity: “Red man” syndrome • nephrotoxicity

  •  Teicoplanin: Intravenous, similar to vancomycin except that long half-life (45–70 h) permits once-daily dosing
  •  Dalbavancin: Intravenous, very long half-life (>10 days) permits once-weekly dosing
  •  Oritavancin: Intravenous, very long half-life (>10 days) permits once-weekly dosing
  •  Telavancin: Intravenous, once-daily dosing

LIPOPEPTIDE
  •  Daptomycin Binds to cell membrane, Bactericidal activity against Infections caused by Gram- IV administration • renal clearance (half-life 8 h)
causing depolarization susceptible bacteria • more positive bacteria including • dosed once daily • inactivated by pulmonary
and rapid cell death rapidly bactericidal than sepsis and endocarditis surfactant so cannot be used to treat pneumonia
vancomycin • Toxicity: Myopathy • monitoring of weekly
creatine phosphokinase levels recommended
CHAPTER 43  Beta-Lactam & Other Cell Wall- & Membrane-Active Antibiotics     813

P R E P A R A T I O N S A V A I L A B L E

GENERIC NAME AVAILABLE AS GENERIC NAME AVAILABLE AS


PENICILLINS Broad-spectrum (third- & fourth-generation) cephalosporins
Amoxicillin Generic, Amoxil, others  Cefdinir Generic
Amoxicillin/potassium Generic, Augmentin   Cefditoren pivoxil Spectracef
clavulanate*  Cefepime Generic, Maxipime
Ampicillin Generic  Cefixime Suprax
Ampicillin/sulbactam sodium† Generic, Unasyn  Cefotaxime Generic, Claforan
Dicloxacillin Generic, Dynapen   Cefpodoxime proxetil Generic
Nafcillin Generic, Nallpen   Ceftaroline fosamil Teflaro
Oxacillin Generic, Bactocill  Ceftazidime Generic, Fortaz, Tazicef
Penicillin G Generic, Pfizerpen Ceftazidime/avibactam§ Avycaz
Penicillin G benzathine Permapen, Bicillin L-A  Ceftibuten Generic, Cedax
Penicillin G procaine Generic Ceftolozane/tazobactam|| Zerbaxa
Penicillin V Generic, V-Cillin, Pen-Vee K,  Ceftriaxone Generic, Rocephin
others Monobactam & Carbapenems
Piperacillin and tazobactam Zosyn  Aztreonam Generic, Azactam, Cayston

sodium
 Doripenem Doribax
CEPHALOSPORINS & OTHER BETA-LACTAM DRUGS
 Ertapenem Invanz
Narrow-spectrum (first-generation) cephalosporins
 Imipenem/cilastatin Generic, Primaxin IM, Primaxin IV
 Cefadroxil Generic
 Meropenem Generic, Merrem IV
 Cefazolin Generic, Ancef, Kefzol
OTHER DRUGS DISCUSSED IN THIS CHAPTER
 Cephalexin Generic, Keflex, others
Cycloserine Generic
Intermediate-spectrum (second-generation) cephalosporins
Dalbavancin Dalvance
 Cefaclor Generic Daptomycin Cubicin
 Cefotetan Generic, Cefotan Fosfomycin Monurol
 Cefoxitin Generic Oritavancin Orbactiv
 Cefprozil Generic Telavancin Vibativ
 Cefuroxime Generic, Ceftin, Zinacef Vancomycin Generic, Vancocin
*
Clavulanate content varies with the formulation; see package insert.

Sulbactam content is half the ampicillin content.

Tazobactam content is 12.5% of the piperacillin content.
§
Avibactam content is 25% of the ceftazidime content.
||
Tazobactam content is half the ceftolozane content.

REFERENCES Corey GR et al: Single-dose oritavancin versus 7-10 days of vancomycin in the
treatment of gram-positive acute bacterial skin and skin structure infections:
Biek D et al: Ceftaroline fosamil: A novel broad-spectrum cephalosporin with The SOLO II noninferiority study. Clin Infect Dis 2015;60:254.
expanded Gram-positive activity. J Antimicrob Chemother 2010;65(Suppl
DePestel DD et al: Cephalosporin use in treatment of patients with penicillin aller-
4):iv9.
gies. J Am Pharm Assoc 2008;48:530.
Billeter M et al: Dalbavancin: A novel once-weekly lipoglycopeptide antibiotic.
Fowler VG et al: Daptomycin versus standard therapy for bacteremia and endocar-
Clin Infect Dis 2008;46:577.
ditis caused by Staphylococcus aureus. N Engl J Med 2006;355:653.
Boucher HW et al: Once-weekly dalbavancin versus daily conventional therapy for
Jacoby GA, Munoz-Price LS: The new beta-lactamases. N Engl J Med
skin infection. N Engl J Med 2014;370:2169.
2005;352:380.
Carpenter CF, Chambers HF: Daptomycin: Another novel agent for treating
Keating GM, Perry CM: Ertapenem: A review of its use in the treatment of bacte-
infections due to drug-resistant gram-positive pathogens. Clin Infect Dis
rial infections. Drugs 2005;65:2151.
2004;38:994.
Kerneis S et al: Cefoxitin as a carbapenem-sparing antibiotic for infections caused
Centers for Disease Control and Prevention (CDC): Antibiotic resistance threats
by extended-spectrum beta-lactamase producing Escherichia coli and Klebsi-
in the United States, 2013. Available at: www.cdc.gov/drugresistance/
ella pneumoniae. Infect Dis 2015;47:789.
threat-report-2013/.
Lee SH et al: TarO-specific inhibitors of wall teichoic acid biosynthesis restore
Chang C et al: Overview of penicillin allergy. Clinic Rev Allerg Immunol
β-lactam efficacy against methicillin-resistant staphylococci. Sci Transl Med
2012;43:84.
2016;8:329.
Chovel-Sella A et al: The incidence of rash after amoxicillin treatment in children
with infectious mononucleosis. Pediatrics 2013;131:1424.
814    SECTION VIII  Chemotherapeutic Drugs

Leonard SN, Rybak MJ: Telavancin: An antimicrobial with a multifunctional Infectious Diseases Society of America, and the Society of Infectious Diseases
mechanism of action for the treatment of serious gram-positive infections. Pharmacists. Am J Health Syst Pharm 2009;66:82.
Pharmacotherapy 2008;28:458. Sievart DM et al: Vancomycin-resistant Staphylococcus aureus in the United
Mandell L: Doripenem: A new carbapenem in the treatment of nosocomial infec- States, 2002-2006. Clin Infect Dis 2008;46:668.
tions. Clin Infect Dis 2009;49(Suppl 1):S1. Tamma PD et al: The use of cefepime for treating AmpC β-lactamase-producing
Marston HD et al: Antimicrobial resistance. JAMA 2016;316:1193. enterobacteriaceae. Clin Infect Dis 2013;57:781.
Noskin GA et al: National trends in Staphylococcus aureus infection rates: Impact Van Duin D and Bonomo RA: Ceftazidime/avibactam and ceftolozane/tazobac-
on economic burden and mortality over a 6-year period. Clin Infect Dis tam: Second-generation β-lactam/β-lactamase combinations. Clin Infect
2007;45:1132. Dis 2016;63;234.
Rybak M et al: Therapeutic monitoring of vancomycin in adult patients: A con- Zar FA et al: A comparison of vancomycin and metronidazole for the treatment of
sensus review of the American Society of Health-System Pharmacists, the Clostridium difficile-associated diarrhea. Clin Infect Dis 2007;45:302.

C ASE STUDY ANSWER

An intravenous third-generation cephalosporin (ceftriaxone patient has a history of rash to amoxicillin, the presentation
or cefotaxime) with adequate penetration into inflamed was not consistent with an anaphylactic reaction. The ami-
meninges that is active against the common bacteria that nopenicillins are frequently associated with rashes that are
cause community-acquired pneumonia and meningitis not caused by Type I hypersensitivity. In this instance, cross-
(pneumococcus, meningococcus, Haemophilus) should be reactivity with a cephalosporin is unlikely—particularly
ordered. Vancomycin also should be administered until with a third-generation drug—and the patient presents with
culture and sensitivity results are available in case the patient life-threatening illness necessitating appropriate and proven
is infected with a resistant pneumococcus. Although the antibiotic coverage.
46
C H A P T E R

Sulfonamides,
Trimethoprim, &
Quinolones
Camille E. Beauduy, PharmD, & Lisa G. Winston, MD*

C ASE STUDY

A 59-year-old woman presents to an urgent care clinic with tract infections in the past year. Each episode was uncom-
a 4-day history of frequent and painful urination. She has plicated, treated with trimethoprim-sulfamethoxazole, and
had fevers, chills, and flank pain for the past 2 days. Her promptly resolved. She also has osteoporosis for which she
physician advised her to come immediately to the clinic for takes a daily calcium supplement. The decision is made to
evaluation. In the clinic she is febrile (38.5°C [101.3°F]) treat her with oral antibiotics for a complicated urinary
but otherwise stable and states she is not experiencing any tract infection with close follow-up. Given her history,
nausea or vomiting. Her urine dipstick test is positive for what would be a reasonable empiric antibiotic choice?
leukocyte esterase. Urinalysis and urine culture are ordered. Depending on the antibiotic choice are there potential drug
Her past medical history is significant for three urinary interactions?

■■ ANTIFOLATE DRUGS Mechanism of Action & Antimicrobial


Activity
SULFONAMIDES Sulfonamide-susceptible organisms, unlike mammals, cannot
use exogenous folate but must synthesize it from PABA. This
Chemistry pathway (Figure 46–2) is thus essential for production of
The basic formulas of the sulfonamides and their structural simi- purines and nucleic acid synthesis. As structural analogs of
larity to p-aminobenzoic acid (PABA) are shown in Figure 46–1. PABA, sulfonamides inhibit dihydropteroate synthase and folate
Sulfonamides with varying physical, chemical, pharmacologic, production. Sulfonamides inhibit both Gram-positive bacteria,
and antibacterial properties are produced by attaching sub- such as Staphylococcus sp and Gram-negative enteric bacteria such
stituents to the amido group ( ⎯  SO2   ⎯  NH   ⎯   R) or the amino as Escherichia coli, Klebsiella pneumoniae, Salmonella, Shigella, and
group ( ⎯  NH2) of the sulfanilamide nucleus. Sulfonamides tend Enterobacter sp, as well as Nocardia sp, Chlamydia trachomatis, and
to be much more soluble at alkaline than at acid pH. Most can some protozoa. Rickettsiae are not inhibited by sulfonamides but
be prepared as sodium salts, which are used for intravenous are instead stimulated in their growth. Activity is poor against
administration. anaerobes. Pseudomonas aeruginosa is intrinsically resistant to
sulfonamide antibiotics.
Combination of a sulfonamide with an inhibitor of dihy-
*
The authors thank Henry F. Chambers, MD and Daniel H. Deck, for drofolate reductase (trimethoprim or pyrimethamine) provides
their contributions to previous editions.

834
CHAPTER 46  Sulfonamides, Trimethoprim, & Quinolones    835

synthase with low sulfonamide affinity is often encoded on a plas-


SO2NH2 COOH mid that is transmissible and can disseminate rapidly and widely.
Sulfonamide-resistant dihydropteroate synthase mutants also can
emerge under selective pressure.

NH2 NH2
Pharmacokinetics
Sulfanilamide p-Aminobenzoic acid (PABA) Sulfonamides can be divided into three major groups: (1)
oral, absorbable; (2) oral, nonabsorbable; and (3) topical.
SO2NH Oral absorbable sulfonamides are absorbed from the stomach
SO2NH and small intestine and distributed widely to tissues and body
N N
N fluids (including the central nervous system and cerebrospinal
O CH3 fluid), placenta, and fetus. Protein binding varies from 20%
to over 90%. Therapeutic concentrations are in the range of
NH2 40–100 mcg/mL of blood. Blood levels generally peak 2–6 hours
NH2 after oral administration.
Sulfadiazine Sulfamethoxazole A portion of absorbed drug is acetylated or glucuronidated in
the liver. Sulfonamides and inactive metabolites are then excreted
FIGURE 46–1  Structures of some sulfonamides and in the urine, mainly by glomerular filtration. The dosage of
p-aminobenzoic acid. sulfonamides must be reduced in patients with significant renal
failure.

synergistic activity because of sequential inhibition of folate


synthesis (Figure 46–2). Clinical Uses
Sulfonamides are infrequently used as single agents. Many
Resistance strains of formerly susceptible species, including meningococci,
pneumococci, streptococci, staphylococci, and gonococci, are
Some bacteria lack the enzymes required for folate synthesis from now resistant. The fixed-drug combination of trimethoprim-
PABA and, like mammals, depend on exogenous sources of folate; sulfamethoxazole is the drug of choice for infections such
therefore, they are not susceptible to sulfonamides. Sulfonamide as Pneumocystis jiroveci (formerly P carinii) pneumonia,
resistance may also occur as a result of mutations that (1) cause toxoplasmosis, and nocardiosis.
overproduction of PABA, (2) cause production of a folic acid-
synthesizing enzyme that has low affinity for sulfonamides, or
A. Oral Absorbable Agents
(3) impair permeability to the sulfonamide. Dihydropteroate
Sulfamethoxazole is a commonly used absorbable agent; however,
in the USA, it is available only as the fixed-dosed combination
trimethoprim-sulfamethoxazole. Typical dosing and indications
are discussed below.
p-Aminobenzoic acid
Administration of sulfadiazine with pyrimethamine is first-
line therapy for treatment of acute toxoplasmosis. Using sulfa-
Sulfonamides
Dihydropteroate − (compete diazine plus pyrimethamine, a potent inhibitor of dihydrofolate
synthase
with PABA) reductase, is synergistic because these drugs block sequential steps
in the folate synthesis pathway (Figure 46–2). However, since
Dihydrofolic acid 2015, there have been challenges with manufacturing, supply,
and pricing of pyrimethamine in the USA. In some cases, clini-
cians have obtained a compounded product through specialty
Dihydrofolate − Trimethoprim pharmacies or prescribed alternate agents, such as trimethoprim-
reductase
sulfamethoxazole. Sulfadoxine is a long-acting sulfonamide that is
coformulated with pyrimethamine (Fansidar). This combination
Tetrahydrofolic acid
is no longer commercially available in the USA but may be found
in other parts of the world where it is used as a second-line treat-
ment for malaria (see Chapter 52).
Purines

B. Oral Nonabsorbable Agents


DNA Sulfasalazine (salicylazosulfapyridine) is widely used in ulcer-
ative colitis, enteritis, and other inflammatory bowel disease (see
FIGURE 46–2  Actions of sulfonamides and trimethoprim. Chapter 62).
836    SECTION VIII  Chemotherapeutic Drugs

C. Topical Agents and sulfamethoxazole is often bactericidal, compared with the


Sodium sulfacetamide ophthalmic solution or ointment is effec- bacteriostatic activity of a sulfonamide alone.
tive in the treatment of bacterial conjunctivitis and as adjunctive NH2 OCH3
therapy for trachoma. Another sulfonamide, mafenide acetate, is N
used topically but can be absorbed from burn sites. The drug and
H2N CH2 OCH3
its primary metabolite inhibit carbonic anhydrase and can cause
metabolic acidosis, a side effect that limits its usefulness. Silver N

sulfadiazine is a less toxic topical sulfonamide and is preferred to OCH3


mafenide for prevention of infection of burn wounds. Trimethoprim

Adverse Reactions NH2


N
Historically, drugs containing a sulfonamide moiety, including
antimicrobial sulfas, diuretics, diazoxide, and the sulfonylurea hypo- H2N CI
glycemic agents, were considered to be cross-allergenic. However, N
more recent evidence suggests cross-reactivity is uncommon and C2H5
many patients who are allergic to nonantibiotic sulfonamides toler- Pyrimethamine
ate sulfonamide antibiotics. The most common adverse effects are
fever, skin rashes, exfoliative dermatitis, photosensitivity, urticaria,
nausea, vomiting, diarrhea, and difficulties referable to the urinary Resistance
tract (see below). Stevens-Johnson syndrome, although relatively Resistance to trimethoprim can result from reduced cell perme-
uncommon (<1% of treatment courses), is a particularly serious ability, overproduction of dihydrofolate reductase, or production
and potentially fatal type of skin and mucous membrane eruption of an altered reductase with reduced drug binding. Resistance
associated with sulfonamide use. Other unwanted effects include can emerge by mutation, although more commonly it is due to
stomatitis, conjunctivitis, arthritis, hematopoietic disturbances (see plasmid-encoded trimethoprim-resistant dihydrofolate reductases.
below), hepatitis, and, rarely, polyarteritis nodosa and psychosis. These resistant enzymes may be coded within transposons on con-
jugative plasmids that exhibit a broad host range, accounting for
A. Urinary Tract Disturbances rapid and widespread dissemination of trimethoprim resistance
Sulfonamides may precipitate in urine, especially at neutral or acid among numerous bacterial species.
pH, producing crystalluria, hematuria, or even obstruction. This is
rarely a problem with the more soluble sulfonamides (eg, sulfisoxa- Pharmacokinetics
zole). Sulfadiazine and sulfamethoxazole are relatively insoluble in Trimethoprim is usually given orally, alone or in combination with
acidic urine and can cause crystalluria, particularly when given in sulfamethoxazole, which has a similar half-life. Trimethoprim-
large doses or if fluid intake is poor. Crystalluria is treated by admin- sulfamethoxazole can also be given intravenously. Trimethoprim is
istration of sodium bicarbonate to alkalinize the urine and fluids well absorbed from the gut and distributed widely in body fluids
to increase urine flow. Sulfonamides have also been implicated in and tissues, including cerebrospinal fluid.
various types of nephrosis and in allergic nephritis. Because trimethoprim is more lipid-soluble than sulfamethoxa-
zole, it has a larger volume of distribution than the latter drug.
B. Hematopoietic Disturbances Therefore, when 1 part of trimethoprim is given with 5 parts of
Sulfonamides can cause hemolytic or aplastic anemia, granulocy- sulfamethoxazole (the ratio in the formulation), the peak plasma con-
topenia, thrombocytopenia, or leukemoid reactions. Sulfonamides centrations are in the ratio of 1:20, which is optimal for the combined
may provoke hemolytic reactions in patients with glucose- effects of these drugs in vitro. About 30–50% of the sulfonamide and
6-phosphate dehydrogenase deficiency. Sulfonamides taken near 50–60% of the trimethoprim (or their respective metabolites) are
the end of pregnancy increase the risk of kernicterus in newborns. excreted in the urine within 24 hours. The dose should be reduced by
half for patients with creatinine clearances of 15–30 mL/min.
Trimethoprim (a weak base) concentrates in prostatic fluid and
TRIMETHOPRIM & TRIMETHOPRIM- in vaginal fluid, which are more acidic than plasma. Therefore, it
SULFAMETHOXAZOLE MIXTURES has more antibacterial activity in prostatic and vaginal fluids than
many other antimicrobial drugs.
Mechanism of Action
Trimethoprim, a trimethoxybenzylpyrimidine, selectively inhib-
Clinical Uses
its bacterial dihydrofolic acid reductase, which converts dihy- A. Oral Trimethoprim
drofolic acid to tetrahydrofolic acid, a step leading to the Trimethoprim can be given alone (100 mg twice daily) in acute
synthesis of purines and ultimately to DNA (Figure 46–2). urinary tract infections. Many community-acquired organisms are
Trimethoprim is a much less efficient inhibitor of mammalian susceptible to the high concentrations that are found in the urine
dihydrofolic acid reductase. The combination of trimethoprim (200–600 mcg/mL).
CHAPTER 46  Sulfonamides, Trimethoprim, & Quinolones    837

B. Oral Trimethoprim-Sulfamethoxazole (TMP-SMZ) acid, 10 mg orally each day, should be administered to minimize
A combination of trimethoprim-sulfamethoxazole is effective bone marrow suppression seen with pyrimethamine. Some clini-
treatment for a wide variety of infections including P jiroveci cians recommend using trimethoprim-sulfamethoxazole as an
pneumonia, urinary tract infections, prostatitis, and some alternate option if pyrimethamine is not available.
infections caused by susceptible strains of Shigella, Salmonella, In falciparum malaria, the combination of pyrimethamine
and nontuberculous mycobacteria. It is active against most with sulfadoxine (Fansidar) has been used (see Chapter 52); how-
Staphylococcus aureus strains, both methicillin-susceptible and ever, it is no longer commercially available in the USA.
methicillin-resistant, and against respiratory tract pathogens such
as Haemophilus sp, Moraxella catarrhalis, and K pneumoniae (but Adverse Effects
not Mycoplasma pneumoniae). However, the increasing prevalence Trimethoprim produces the predictable adverse effects of an
of strains of E coli (up to 30% or more) and pneumococci that are antifolate drug, especially megaloblastic anemia, leukopenia, and
resistant to trimethoprim-sulfamethoxazole must be considered granulocytopenia. The combination trimethoprim-sulfamethox-
before using this combination for empiric therapy of upper uri- azole may cause all of the untoward reactions associated with
nary tract infections or pneumonia. Trimethoprim-sulfamethoxa- sulfonamides. Nausea and vomiting, drug fever, vasculitis, renal
zole is commonly used for the treatment of uncomplicated skin damage, and central nervous system disturbances occasionally
and soft tissue infections. occur. Patients with AIDS and pneumocystis pneumonia have
One double-strength tablet (each tablet contains trimethoprim a particularly high frequency of untoward reactions to trime-
160 mg plus sulfamethoxazole 800 mg) given every 12 hours is thoprim-sulfamethoxazole, especially fever, rashes, leukopenia,
effective treatment for urinary tract infections, prostatitis, uncom- diarrhea, elevations of hepatic aminotransferases, hyperkalemia,
plicated skin and soft tissue infections, and infections caused by and hyponatremia. Trimethoprim inhibits secretion of creatinine
susceptible strains of Shigella and Salmonella. Bone and joint at the distal renal tubule, resulting in mild elevation of serum
infections caused by S. aureus can be effectively treated, typically creatinine without impairment of glomerular filtration rate. This
at doses of 8–10 mg/kg per day of the trimethoprim component. nontoxic effect is important to distinguish from true nephrotoxic-
One single-strength tablet (containing trimethoprim 80 mg plus ity that may be caused by sulfonamides.
sulfamethoxazole 400 mg) given three times weekly may serve as
prophylaxis in recurrent urinary tract infections of some women.
The dosage for children treated for shigellosis, urinary tract infec-
tion, or otitis media is trimethoprim 8 mg/kg per day and sulfa-
■■ DNA GYRASE INHIBITORS
methoxazole 40 mg/kg per day divided every 12 hours.
Infections with P jiroveci and some other pathogens, such as
FLUOROQUINOLONES
Nocardia or Stenotrophomonas maltophilia, can be treated with high The clinically relevant quinolones are synthetic fluorinated ana-
doses of the either the oral or intravenous combination (dosed on logs of nalidixic acid (Figure 46–3). They are active against a
the basis of the trimethoprim component at 15–20 mg/kg/d). variety of Gram-positive and Gram-negative bacteria.
P jiroveci can be prevented in immunosuppressed patients by a
number of low dose regimens such as one double-strength tablet
daily or three times weekly. Mechanism of Action
Quinolones block bacterial DNA synthesis by inhibiting bacterial
C. Intravenous Trimethoprim-Sulfamethoxazole topoisomerase II (DNA gyrase) and topoisomerase IV. Inhibition
A solution of the mixture containing 80 mg trimethoprim plus of DNA gyrase prevents the relaxation of positively supercoiled
400 mg sulfamethoxazole per 5 mL diluted in 125 mL of 5% DNA that is required for normal transcription and replication.
dextrose in water can be administered by intravenous infusion Inhibition of topoisomerase IV interferes with separation of repli-
over 60–90 minutes. It is the agent of choice for moderately cated chromosomal DNA into the respective daughter cells during
severe to severe pneumocystis pneumonia. It has been used for cell division.
Gram-negative bacterial sepsis, but has largely been replaced by
extended spectrum β-lactams and fluoroquinolones. It may be Antibacterial Activity
an effective alternative for infections caused by some multidrug- Earlier quinolones such as nalidixic acid did not achieve systemic
resistant species such as Enterobacter and Serratia; shigellosis; or antibacterial levels and were useful only in the treatment of lower
typhoid. It is the preferred alternate therapy for serious Listeria urinary tract infections. Fluorinated derivatives (ciprofloxacin,
infections in patients unable to tolerate ampicillin. The dosage is levofloxacin, and others; Figure 46–3 and Table 46–1) have
10–20 mg/kg/d of the trimethoprim component. greatly improved antibacterial activity compared with nalidixic
acid and achieve bactericidal levels in blood and tissues.
D. Oral Pyrimethamine with Sulfonamide Fluoroquinolones were originally developed because of their
Pyrimethamine and sulfadiazine are used in the treatment of toxo- excellent activity against Gram-negative aerobic bacteria; the ear-
plasmosis. The dosage of sulfadiazine is 1–1.5 g four times daily, liest agents had limited activity against Gram-positive organisms.
with pyrimethamine given as a 200-mg loading dose followed by Subsequent members of the group have improved activity against
a once-daily dose of 50–75 mg. Leucovorin, also known as folinic Gram-positive cocci. The relative activity against Gram-negative
838    SECTION VIII  Chemotherapeutic Drugs

O
O
COOH F COOH

CH3 N N HN N N
C2H5
C2H5
Nalidixic acid Norfloxacin

O O
F COOH F COOH

HN N N CH3 N N N
O
CH3
Ciprofloxacin Levofloxacin

O
O
F COOH
CH2O F CO2H
H N N
N N
N N N
O
H3C H2N

Moxifloxacin Gemifloxacin

FIGURE 46–3  Structures of nalidixic acid and some fluoroquinolones.

versus Gram-positive species is useful for differentiating these typically used in combination with a second active agent, such
agents. Norfloxacin, which is no longer available in the USA, as rifampin, to prevent emergence of resistance while on therapy.
is the least active of the fluoroquinolones against both Gram- Enterococci tend to be less susceptible than staphylococci, limit-
negative and Gram-positive organisms, with minimum inhibitory ing the efficacy of fluoroquinolones in infections caused by these
concentrations (MICs) fourfold to eightfold higher than those organisms. Ciprofloxacin is the most active agent of this group
of ciprofloxacin. Ciprofloxacin, enoxacin, lomefloxacin, levo- against Gram-negative organisms, particularly P aeruginosa. Levo-
floxacin, ofloxacin, and pefloxacin comprise a second group floxacin, the l-isomer of ofloxacin, has superior activity against
of similar agents possessing excellent Gram-negative activity and Gram-positive organisms, especially Streptococcus pneumoniae.
moderate to good activity against Gram-positive bacteria. Cip- Gatifloxacin, gemifloxacin, and moxifloxacin make up a third
rofloxacin and levofloxacin are the two agents from this group group of fluoroquinolones with improved activity against Gram-
that are used systemically in the USA. MICs for Gram-negative positive organisms, particularly S pneumoniae and some staphylo-
cocci and bacilli, including Enterobacter sp, P aeruginosa, Neis- cocci. Gemifloxacin is active in vitro against ciprofloxacin-resistant
seria meningitidis, Haemophilus sp, and Campylobacter jejuni, are strains of S pneumoniae, but in vivo efficacy is unproven. Although
1–2 mcg/mL and often less. Methicillin-susceptible strains of S MICs of these agents for staphylococci are lower than those of cip-
aureus are generally susceptible to these fluoroquinolones, but rofloxacin (and the other compounds mentioned in the paragraph
methicillin-resistant strains of staphylococci are often resistant. above), it is not known whether the enhanced activity is sufficient
When treating staphylococcal infections, fluoroquinolones are to permit use of these agents for treatment of infections caused by

TABLE 46–1  Pharmacokinetic properties of some fluoroquinolones.


Oral Peak Serum Primary Route of
Drug Half-Life (h) Bioavailability (%) Concentration (mcg/mL) Oral Dose (mg) Excretion

Ciprofloxacin 3–5 70 2.4 500 twice daily Renal


Gemifloxacin 8 70 1.6 320 once daily Renal and nonrenal
Levofloxacin 5–7 95 5.7 500 once daily Renal
Moxifloxacin 9–10 >85 3.1 400 once daily Nonrenal
Norfloxacin 3.5–5 80 1.5 400 twice daily Renal
Ofloxacin 5–7 95 2.9 400 twice daily Renal
CHAPTER 46  Sulfonamides, Trimethoprim, & Quinolones    839

ciprofloxacin-resistant strains. In general, none of these agents is as agents are also effective for bacterial diarrhea caused by Shigella,
active as ciprofloxacin against Gram-negative organisms. Fluoro- Salmonella, toxigenic E coli, and Campylobacter. Fluoroquinolones
quinolones also are active against agents of atypical pneumonia (eg, (except norfloxacin, which does not achieve adequate systemic
mycoplasmas and chlamydiae) and against intracellular pathogens concentrations) are used in infections of soft tissues, bones, and
such as Legionella and some mycobacteria, including Mycobacterium joints and in intra-abdominal and respiratory tract infections,
tuberculosis and Mycobacterium avium complex. Moxifloxacin has including those caused by multidrug-resistant organisms such as
modest activity against anaerobic bacteria but lacks appreciable Pseudomonas and Enterobacter. Ciprofloxacin is a drug of choice
activity against P aeruginosa. Because of toxicity when systemically for prophylaxis and treatment of anthrax; the newer fluoroquino-
administered, gatifloxacin is available only as an ophthalmic solu- lones are active in vitro, and levofloxacin is also approved by the
tion in the USA. U.S. Food and Drug Administration (FDA) for prophylaxis.
Ciprofloxacin and levofloxacin are no longer recommended
Resistance for the treatment of gonococcal infection in the USA, as resis-
tance is now common; however, gemifloxacin may be used in
During fluoroquinolone therapy, resistant organisms emerge in combination with azithromycin as an alternate to ceftriaxone.
about 1 of every 107–109 organisms, especially among staphylo- Levofloxacin and ofloxacin are recommended by the Centers for
cocci, P aeruginosa, and Serratia marcescens. Emerging resistance Disease Control and Prevention as alternative treatment options
is due to one or more point mutations in the quinolone binding for chlamydial urethritis or cervicitis. Ciprofloxacin, levofloxacin,
region of the target enzyme or to a change in the permeability of or moxifloxacin is occasionally used as part of a treatment regimen
the organism. However, additional mechanisms seem to account for tuberculosis and non-tuberculous mycobacterial infections.
for the relative ease with which resistance develops in highly sus- These agents are suitable for eradication of meningococci from
ceptible bacteria. Two types of plasmid-mediated resistance have carriers and for prophylaxis of bacterial infection in neutropenic
been described. The first type utilizes Qnr proteins, which protect cancer patients.
DNA gyrase from the fluoroquinolones. The second is a variant With their enhanced Gram-positive activity and activity
of an aminoglycoside acetyltransferase capable of modifying cip- against atypical pneumonia agents (chlamydiae, Mycoplasma,
rofloxacin. Both mechanisms confer low-level resistance that may and Legionella), levofloxacin, gemifloxacin, and moxifloxacin—
facilitate the point mutations that confer high-level resistance and so-called respiratory fluoroquinolones—are effective for treatment
also may be associated with resistance to other antibacterial drug of lower respiratory tract infections.
classes. Resistance to one fluoroquinolone, particularly if it is of
high level, generally confers cross-resistance to all other members
of this class. Adverse Effects
Fluoroquinolones are generally well tolerated. The most common
Pharmacokinetics effects are nausea, vomiting, and diarrhea. Occasionally, headache,
After oral administration, the fluoroquinolones are well absorbed dizziness, insomnia, skin rash, or abnormal liver function tests
(bioavailability of 80–95%) and distributed widely in body fluids develop. Photosensitivity has been reported with lomefloxacin
and tissues (Table 46–1). Serum half-lives range from 3 to 10 and pefloxacin. Prolongation of the QTc interval may occur with
hours. The relatively long half-lives of levofloxacin, gemifloxacin, gatifloxacin, levofloxacin, gemifloxacin, and moxifloxacin; these
and moxifloxacin permit once-daily dosing. Oral absorption is drugs should be avoided or used with caution in patients with
impaired by divalent and trivalent cations, including those in ant- known QTc interval prolongation or uncorrected hypokalemia;
acids. Therefore, oral fluoroquinolones should be taken 2 hours in those receiving class 1A (eg, quinidine or procainamide) or
before or 4 hours after any products containing these cations. class 3 antiarrhythmic agents (sotalol, ibutilide, amiodarone);
Serum concentrations of intravenously administered drug are sim- and in patients receiving other agents known to increase the QTc
ilar to those of orally administered drug. Most fluoroquinolones, interval (eg, erythromycin, tricyclic antidepressants). Gatifloxacin
moxifloxacin being an important exception, are eliminated by has been associated with hyperglycemia in diabetic patients and
renal mechanisms, either tubular secretion or glomerular filtration with hypoglycemia in patients also receiving oral hypoglycemic
(Table 46–1). Dosage adjustment is required for patients with agents. Because of these serious effects (including some fatalities),
creatinine clearances less than 50 mL/min, the exact adjustment gatifloxacin was withdrawn from sale in the United States in 2006.
depending on the degree of renal impairment and the specific In animal models, fluoroquinolones may damage growing car-
fluoroquinolone being used. Dosage adjustment for renal failure is tilage and cause an arthropathy. Thus, these drugs have not been
not necessary for moxifloxacin since it is metabolized in the liver; recommended as first-line agents for patients under 18 years of
it should be used with caution in patients with hepatic failure. age. However, there is a growing consensus that fluoroquinolones
may be used in children if needed (eg, for treatment of pseu-
domonal infections in patients with cystic fibrosis). Tendinitis,
Clinical Uses a complication in adults, can be serious because of the risk of
Fluoroquinolones (other than moxifloxacin, which achieves rela- tendon rupture. Risk factors for tendinitis include advanced age,
tively low urinary levels) are effective in urinary tract infec- renal insufficiency, and concurrent steroid use. Fluoroquinolones
tions caused by many organisms, including P aeruginosa. These should be avoided during pregnancy in the absence of specific
840    SECTION VIII  Chemotherapeutic Drugs

data documenting their safety. Oral or intravenously adminis- many potential adverse effects are uncommon, the FDA called for
tered fluoroquinolones have also been associated with peripheral updated warnings for all fluoroquinolones in 2016, stating that
neuropathy. Neuropathy can occur at any time during treatment these agents should be reserved for patients who do not have alter-
with fluoroquinolones and may persist for months to years after native options, particularly in less severe infections such as upper
the drug is stopped. In some cases it may be permanent. Although respiratory infections or uncomplicated cystitis.

SUMMARY Sulfonamides, Trimethoprim, and Fluoroquinolones


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

FOLATE ANTAGONISTS
  • Trimethoprim- Synergistic combination of Bactericidal activity Urinary tract infections • soft Oral, IV • renal clearance (half-life 8 h)
sulfamethoxazole folate antagonists blocks against susceptible tissue infections • bone and • dosed every 8–12 h • formulated in a
purine production and bacteria joint infections • P jiroveci 5:1 ratio of sulfamethoxazole to
nucleic acid synthesis pneumonia • toxoplasmosis trimethoprim • Toxicity: Rash, fever, bone
• nocardiosis marrow suppression, hyperkalemia,
nephrotoxicity

  •  Sulfadiazine: Oral; first-line therapy for toxoplasmosis when combined with pyrimethamine
  •  Trimethoprim: Oral; used alone only for lower urinary tract infections; may be safely prescribed to patients with sulfonamide allergy
  •  Pyrimethamine: Oral; first-line therapy for toxoplasmosis when combined with sulfadiazine; coadminister with leucovorin to limit bone marrow toxicity
  •  Pyrimethamine-sulfadoxine: Oral; second-line malaria treatment

FLUOROQUINOLONES
  •  Ciprofloxacin Inhibits DNA replication by Bactericidal activity Urinary tract infections Oral, IV • mixed clearance (half-life 4 h)
binding to DNA gyrase and against susceptible • gastroenteritis • dosed every 12 h • divalent and trivalent
topoisomerase IV bacteria • osteomyelitis • anthrax cations impair oral absorption • Toxicity:
Gastrointestinal upset, neurotoxicity,
tendonitis

  • Levofloxacin: Oral, IV; l-isomer of ofloxacin; once-daily dosing; renal clearance; “respiratory” fluoroquinolone with improved activity versus pneumococcus
  • Moxifloxacin: Oral, IV; “respiratory” fluoroquinolone; once-daily dosing; improved activity versus anaerobes and M tuberculosis; hepatic clearance results in lower urinary
levels so use in urinary tract infections is not recommended
  •  Gemifloxacin: Oral; “respiratory” fluoroquinolone

P R E P A R A T I O N S A V A I L A B L E

GENERIC NAME AVAILABLE AS GENERIC NAME AVAILABLE AS


GENERAL-PURPOSE SULFONAMIDES PYRIMETHAMINE
Sulfadiazine Generic Pyrimethamine Daraprim
SULFONAMIDES FOR SPECIAL APPLICATIONS Pyrimethamine-sulfadoxine Generic, Fansidar
Mafenide Generic, Sulfamylon QUINOLONES & FLUOROQUINOLONES
Silver sulfadiazine Generic, Silvadene Ciprofloxacin Generic, Cipro, Cipro I.V., Ciloxan
Sulfacetamide sodium Generic (ophthalmic)
(ophthalmic) Gemifloxacin Factive
TRIMETHOPRIM Levofloxacin Levaquin, Quixin (ophthalmic)
Trimethoprim Generic, Proloprim, Moxifloxacin Generic, Avelox, others
Trimpex Norfloxacin Noroxin
Trimethoprim-sulfamethoxazole Generic, Bactrim, Septra, others Ofloxacin Generic, Floxin, Ocuflox
(co-trimoxazole, TMP-SMZ) (ophthalmic), Floxin Otic (otic)
CHAPTER 46  Sulfonamides, Trimethoprim, & Quinolones    841

REFERENCES trial of trimethoprim-sulfamethoxazole versus ciprofloxacin. Clin Infect Dis


2010;51:143.
Briasoulis A et al: QT prolongation and torsade de pointes induced by fluoroquino-
Rodriguez-Martinez JM et al: Plasmid-mediated quinolone resistance: An update.
lones: Infrequent side effects from commonly used medications. Cardiology
J Infect Chemother 2011;17:149.
2011;120:103.
Scheld WM: Maintaining fluoroquinolone class efficacy: Review of influencing
Cohen JS: Peripheral neuropathy associated with fluoroquinolones. Ann Pharma-
factors. Emerg Infect Dis 2003;9:1.
cother 2001;35:1540.
Schmitz GR et al: Randomized controlled trial of trimethoprim-sulfamethoxazole
Davidson R et al: Resistance to levofloxacin and failure of treatment of pneumo-
for uncomplicated skin abscesses in patients at risk for community-associated
coccal pneumonia. N Engl J Med 2002;346:747.
methicillin-resistant Staphylococcus aureus infection. Ann Emerg Med
Gupta K et al: International clinical practice guidelines for the treatment of 2010;56:283.
acute uncomplicated cystitis and pyelonephritis in women. Clin Infect Dis
Strom BL et al: Absence of cross-reactivity between sulfonamide antibiotics and
2011;52:103.
sulfonamide nonantibiotics. N Engl J Med 2003;349:1628.
Keating GM, Scott LJ: Moxifloxacin: A review of its use in the management of
Talan DA et al: Prevalence of and risk factor analysis of trimethoprim-sulfamethox-
bacterial infections. Drugs 2004;64:2347.
azole- and fluoroquinolone-resistant E. coli infection among emergency
Mandell LA et al: Infectious Disease Society of America/American Thoracic department patients with pyelonephritis. Clin Infect Dis 2008;47:1150.
Society consensus guidelines on the management of community-acquired
Workowski KA et al: Sexually Transmitted Diseases Treatment Guidelines, 2015.
pneumonia. Clin Infect Dis 2007;44:S27.
MMWR Recomm Rep 2015;64(RR-03):1.
Mwenya DM et al: Impact of cotrimoxazole on carriage and antibiotic resistance
Ziganshina LE et al: Fluoroquinolones for treating tuberculosis (presumed drug
of Streptococcus pneumoniae and Haemophilus influenzae in HIV-infected
sensitive). Cochrane Database Syst Rev 2013;(6):CD004795.
children in Zambia. Antimicrob Agents Chemother 2010;54:3756.
Nouira S et al: Standard versus newer antibacterial agents in the treatment of severe
acute exacerbation of chronic obstructive pulmonary disease: A randomized

C ASE STUDY ANSWER

A fluoroquinolone that achieves good urinary and systemic susceptibility. Her recent exposure to multiple courses of
levels (ciprofloxacin or levofloxacin) would be a reasonable trimethoprim-sulfamethoxazole increases her chances of
choice for empiric treatment of this patient’s complicated having a urinary tract infection with an isolate that is resis-
urinary tract infection. Given the possibility of a fluoroqui- tant to this antibiotic. The patient should be told to take the
nolone-resistant organism, one dose of a parenteral agent oral fluoroquinolone 2 hours before or 4 hours after her
such as ceftriaxone (given IV or IM) would be reason- calcium supplement, as divalent and trivalent cations can
able pending culture results confirming fluoroquinolone significantly impair the absorption of oral fluoroquinolones.
47
C H A P T E R

Antimycobacterial Drugs
Camille E. Beauduy, PharmD, &
Lisa G. Winston, MD*

C ASE STUDY

A 60-year-old man presents to the emergency department tuberculosis, the patient is placed in respiratory isolation.
with a 2-month history of fatigue, weight loss (10 kg), fevers, His first sputum smear shows many acid-fast bacilli, and an
night sweats, and a productive cough. He is currently living HIV test returns with a positive result. What drugs should
with friends and has been intermittently homeless, spending be started for treatment of presumptive pulmonary tubercu-
time in shelters. He reports drinking about 6 beers per day. losis? Does the patient have a heightened risk of developing
In the emergency department, a chest x-ray shows a right medication toxicity? If so, which medication(s) would be
apical infiltrate. Given the high suspicion for pulmonary likely to cause toxicity?

Mycobacteria are intrinsically resistant to most antibiotics. active drugs. An isoniazid-rifampin combination administered for
Because they grow more slowly than other bacteria, antibiotics 9 months will cure 95–98% of cases of tuberculosis caused by sus-
that are most active against rapidly growing cells are relatively ceptible strains. An initial intensive phase of treatment is recom-
ineffective. Mycobacterial cells can also be dormant and, thus, mended for the first 2 months due to the prevalence of resistant
resistant to many drugs or killed only very slowly. The lipid-rich strains. The addition of pyrazinamide during this intensive phase
mycobacterial cell wall is impermeable to many agents. Mycobac- allows the total duration of therapy to be reduced to 6 months
terial species are intracellular pathogens, and organisms residing without loss of efficacy. In practice, therapy is usually initiated
within macrophages are inaccessible to drugs that penetrate these with a four-drug regimen of isoniazid, rifampin, pyrazinamide,
cells poorly. Finally, mycobacteria are notorious for their ability and ethambutol until susceptibility of the clinical isolate has
to develop resistance. Combinations of two or more drugs are been determined. In susceptible isolates, the continuation phase
required to overcome these obstacles and to prevent emergence of consists of an additional 4 months with isoniazid and rifampin
resistance during the course of therapy. The response of mycobac- (Table 47–2). Neither ethambutol nor other drugs such as strep-
terial infections to chemotherapy is slow, and treatment must be tomycin adds substantially to the overall activity of the regimen
administered for months to years, depending on which drugs are (ie, the duration of treatment cannot be further reduced if another
used. The drugs used to treat tuberculosis, atypical mycobacterial drug is used), but the fourth drug provides additional cover-
infections, and leprosy are described in this chapter. age if the isolate proves to be resistant to isoniazid, rifampin,
or both. If therapy is initiated after the isolate is known to be
susceptible to isoniazid and rifampin, ethambutol does not
■■ DRUGS USED IN TUBERCULOSIS need to be added. The prevalence of isoniazid resistance among
clinical isolates in the USA is approximately 10%. Prevalence
Isoniazid (INH), rifampin (or other rifamycin), pyrazinamide,
of resistance to both isoniazid and rifampin (which is termed
and ethambutol are the traditional first-line agents for treatment
multidrug resistance) ranged from 1 to 1.6% from the years
of tuberculosis (Table 47–1). Isoniazid and rifampin are the most
2000 to 2013 in the USA. Multidrug resistance is much more
prevalent in many other parts of the world. Resistance to
*
The authors thank Henry F. Chambers, MD and Daniel H. Deck, rifampin alone is rare.
PharmD for their contributions to previous editions.

842
CHAPTER 47  Antimycobacterial Drugs    843

TABLE 47–1  Antimicrobials used in the treatment of growing tubercle bacilli. It is less effective against nontuberculous
tuberculosis. mycobacteria. Isoniazid penetrates into macrophages and is active
against both extracellular and intracellular organisms.
Drug Typical Adult Dosage1

First-line agents Mechanism of Action & Basis of Resistance


 Isoniazid 300 mg/d Isoniazid inhibits synthesis of mycolic acids, which are essential
 Rifampin 600 mg/d components of mycobacterial cell walls. Isoniazid is a prodrug that
 Pyrazinamide 25 mg/kg/d is activated by KatG, the mycobacterial catalase-peroxidase. The
 Ethambutol 15–25 mg/kg/d activated form of isoniazid forms a covalent complex with an acyl
carrier protein (AcpM) and KasA, a beta-ketoacyl carrier protein
Second-line agents
synthetase, which blocks mycolic acid synthesis. Resistance to
 Amikacin 15 mg/kg/d
isoniazid is associated with mutations resulting in overexpression
  Aminosalicylic acid 8–12 g/d of inhA, which encodes an NADH-dependent acyl carrier pro-
 Bedaquiline 400 mg/d tein reductase; mutation or deletion of the katG gene; promoter
 Capreomycin 15 mg/kg/d mutations resulting in overexpression of ahpC, a gene involved
 Clofazimine 200 mg/d in protection of the cell from oxidative stress; and mutations in
kasA. Overproducers of inhA express low-level isoniazid resis-
 Cycloserine 500–1000 mg/d, divided
tance and cross-resistance to ethionamide. KatG mutants express
 Ethionamide 500–750 mg/d
high-level isoniazid resistance and often are not cross-resistant to
 Levofloxacin 500–750 mg/d ethionamide.
 Linezolid 600 mg/d Drug-resistant mutants are normally present in susceptible
 Moxifloxacin 400 mg/d mycobacterial populations at about 1 bacillus in 106. Since
 Rifabutin2 300 mg/d tuberculous lesions often contain more than 108 tubercle bacilli,
3 resistant mutants are readily selected if isoniazid or any other
 Rifapentine 600 mg once weekly
drug is given as a single agent. The use of two independently
 Streptomycin 15 mg/kg/d
acting drugs in combination is much more effective. The
1
Assuming normal renal function. probability that a bacillus is initially resistant to both drugs is
2
150 mg/d if used concurrently with a protease inhibitor or cobicistat; 600 mg/d with approximately 1 in 106 × 106, or 1 in 1012, several orders of
efavirenz.
3
magnitude greater than the number of infecting organisms.
No longer recommended, but may be considered in selected cases if HIV-uninfected
without cavitation on chest radiograph. Thus, at least two (or more in certain cases) active agents should
always be used to treat active tuberculosis to prevent emergence
of resistance during therapy.
ISONIAZID
Pharmacokinetics
Isoniazid is the most active drug for the treatment of tuberculosis
Isoniazid is readily absorbed from the gastrointestinal tract,
caused by susceptible strains. It is a small molecule (molecular
optimally on an empty stomach; peak concentrations may be
weight 137) that is freely soluble in water. The structural similarity
decreased by up to 50% when taken with a fatty meal. A 300 mg
to pyridoxine is shown below.
oral dose (5 mg/kg in children) achieves peak plasma concentra-
N tions of 3–5 mcg/mL within 1–2 hours. Isoniazid diffuses readily
into all body fluids and tissues. The concentration in the central
nervous system and cerebrospinal fluid ranges between 20% and
100% of simultaneous serum concentrations.
CONHNH2 Metabolism of isoniazid, especially acetylation by liver
Isoniazid
N-acetyltransferase, is genetically determined (see Chapter 4).
The average plasma concentration of isoniazid in rapid acetyl-
ators is about one third to one half of that in slow acetylators,
N and average half-lives are less than 1 hour and 3 hours, respec-
CH3
tively. More rapid clearance of isoniazid by rapid acetylators is
OH usually of no therapeutic consequence when appropriate doses
HOH2C
are administered daily, but subtherapeutic concentrations may
CH2OH occur if drug is administered as a once-weekly dose or if there is
Pyridoxine malabsorption.
Isoniazid metabolites and a small amount of unchanged drug
In vitro, isoniazid inhibits most tubercle bacilli at a concen- are excreted in the urine. The dosage need not be adjusted in renal
tration of 0.2 mcg/mL or less and is bactericidal for actively failure. Dose adjustment is not well defined in patients with severe
844    SECTION VIII  Chemotherapeutic Drugs

TABLE 47–2  Recommended treatment for drug-susceptible tuberculosis.


Intensive Phase Continuation Phase
Regimen (min duration = 8 weeks) (min duration = 18 weeks)1  
(in order of
preference) Drugs Dosing Interval Drugs Dosing Interval Comments
2 2
1 INH 7 days per week INH 7 days per week Preferred regimen.
RIF RIF
PZA
EMB
2 INH 7 days per week2 INH 3 days per week Preferred alternative if less frequent DOT is
RIF RIF needed.
PZA
EMB
3 INH 3 days per week INH 3 days per week Caution in patients with HIV and/or cavitary
RIF RIF disease due to concerns for treatment
failure, relapse, drug resistance.
PZA
EMB
4 INH 7 days per week × INH 2 days per week Avoid in patients with HIV or those with
RIF 2 weeks, then RIF smear-positive and/ or cavitary disease.
PZA 2 days per week ×
EMB 6 weeks
1
Experts recommend prolonged continuation phase (31 weeks) for patients with cavitation on initial chest radiograph and positive cultures at the end of the intensive
treatment phase.
2
May consider 5 days per week if needed for DOT. No studies compare 5 versus 7 doses per week, but extensive experience suggests efficacy of this regimen.
DOT, directly observed therapy; EMB, ethambutol; HIV, human immunodeficiency virus; INH, isoniazid; PZA, pyrazinamide; RIF, rifampin.

preexisting hepatic insufficiency and should be guided by serum A. Immunologic Reactions


concentrations if a reduction in dose is contemplated. Isoniazid Fever and skin rashes are occasionally seen. Drug-induced sys-
inhibits several cytochrome P450 enzymes, leading to increased temic lupus erythematosus has been reported.
concentrations of such medications as phenytoin, carbamazepine,
and benzodiazepines. However, when used in combination with B. Direct Toxicity
rifampin, a potent CYP enzyme inducer, the concentrations of
Isoniazid-induced hepatitis is the most common major toxic effect.
these medications are usually decreased.
This is distinct from the minor increases in liver aminotransferases
(up to three or four times normal), which do not require cessa-
Clinical Uses tion of the drug and which are seen in 10–20% of patients, who
The typical dosage of isoniazid is 5 mg/kg/d; a typical adult dose is usually are asymptomatic. Clinical hepatitis with loss of appetite,
300 mg given once daily. Up to 10 mg/kg/d may be used for seri- nausea, vomiting, jaundice, and right upper quadrant pain occurs
ous infections or if malabsorption is a problem. A 15-mg/kg dose, in 1% of isoniazid recipients and can be fatal, particularly if the
or 900 mg, may be used in a twice to three times-weekly dosing drug is not discontinued promptly. There is histologic evidence of
regimen in combination with a second antituberculous agent (eg, hepatocellular damage and necrosis. The risk of hepatitis depends
rifampin, 600 mg). Pyridoxine, 25–50 mg/d, is recommended on age. It occurs rarely under age 20, in 0.3% of those age 21–35,
for those with conditions predisposing to neuropathy, an adverse 1.2% of those age 36–50, and 2.3% for those age 50 and above.
effect of isoniazid. Isoniazid is usually given by mouth but can be The risk of hepatitis is greater in individuals with alcohol depen-
given parenterally in the same dosage. dence and possibly during pregnancy and the postpartum period.
Isoniazid as a single agent is also indicated for treatment Development of isoniazid hepatitis contraindicates further use of
of latent tuberculosis. The dosage is 300 mg/d (5 mg/kg/d) or the drug.
900 mg twice weekly, and the duration is usually 9 months. Peripheral neuropathy is observed in 10–20% of patients given
dosages greater than 5 mg/kg/d, but it is infrequently seen with
the standard 300-mg adult dose. Peripheral neuropathy is more
Adverse Reactions likely to occur in slow acetylators and patients with predisposing
The incidence and severity of untoward reactions to isoniazid are conditions such as malnutrition, alcoholism, diabetes, AIDS, and
related to dosage and duration of administration. uremia. Neuropathy is due to a relative pyridoxine deficiency.
CHAPTER 47  Antimycobacterial Drugs    845

Isoniazid promotes excretion of pyridoxine, and this toxicity is (see Chapters 4 and 66). Co-administration of rifampin results in
readily reversed by administration of pyridoxine in a dosage as low significantly lower serum levels of these drugs.
as 10 mg/d. Central nervous system toxicity, which is less com-
mon, includes memory loss, psychosis, ataxia, and seizures. These Clinical Uses
effects may also respond to pyridoxine.
A. Mycobacterial Infections
Miscellaneous other reactions include hematologic abnormali-
ties, provocation of pyridoxine deficiency anemia, tinnitus, and Rifampin, usually 600 mg/d (10 mg/kg/d) orally, must be
gastrointestinal discomfort. administered with isoniazid or other antituberculous drugs to
patients with active tuberculosis to prevent emergence of drug-
resistant mycobacteria. In some short-course therapies, 600 mg
of rifampin is given twice weekly. Rifampin, 600 mg daily or
RIFAMPIN
twice weekly for 6 months, also is effective in combination with
Rifampin is a semisynthetic derivative of rifamycin, an antibi- other agents in some atypical mycobacterial infections and in
otic produced by Amycolatopsis rifamycinica, formerly named leprosy. Rifampin, 600 mg daily for 4 months as a single drug,
Streptomyces mediterranei. It is active in vitro against Gram-positive is an alternative to isoniazid for patients with latent tuberculosis
organisms, some Gram-negative organisms, such as Neisseria and who are unable to take isoniazid or who have had exposure to
Haemophilus species, mycobacteria, and chlamydiae. Susceptible a case of active tuberculosis caused by an isoniazid-resistant,
organisms are inhibited by less than 1 mcg/mL. Resistant mutants rifampin-susceptible strain.
are present in all microbial populations at approximately 1 in
106 organisms and are rapidly selected out if rifampin is used as B. Other Indications
a single drug, especially in a patient with active infection. There Rifampin has other uses in bacterial infections. An oral dosage
is no cross-resistance to other classes of antimicrobial drugs, but of 600 mg twice daily for 2 days can eliminate meningococcal
there is cross-resistance to other rifamycin derivatives, eg, rifabutin carriage. Rifampin, 20 mg/kg (maximum 600 mg) once daily
and rifapentine. for 4 days, is used as prophylaxis in contacts of children with
Haemophilus influenzae type b disease. Rifampin combined
Mechanism of Action, Resistance, & with a second agent is sometimes used to eradicate staphy-
lococcal carriage. Rifampin combination therapy is also used
Pharmacokinetics for treatment of serious staphylococcal infections such as
Rifampin binds to the β subunit of bacterial DNA-dependent osteomyelitis, prosthetic joint infections, and prosthetic valve
RNA polymerase and thereby inhibits RNA synthesis. Resistance endocarditis.
results from any one of several possible point mutations in rpoB,
the gene for the β subunit of RNA polymerase. These muta-
tions result in reduced binding of rifampin to RNA polymerase. Adverse Reactions
Human RNA polymerase does not bind rifampin and is not Rifampin imparts a harmless orange color to urine, sweat, and
inhibited by it. Rifampin is bactericidal for mycobacteria. It read- tears (soft contact lenses may be permanently stained). Occasional
ily penetrates most tissues and penetrates into phagocytic cells. It adverse effects include rashes, thrombocytopenia, and nephritis.
can kill organisms that are poorly accessible to many other drugs, Rifampin may cause cholestatic jaundice and occasionally hepa-
such as intracellular organisms and those sequestered in abscesses titis, and it commonly causes light-chain proteinuria. If adminis-
and lung cavities. tered less often than twice weekly, rifampin may cause a flu-like
Rifampin is well absorbed after oral administration and syndrome characterized by fever, chills, myalgias, anemia, and
excreted mainly through the liver into bile. It then undergoes thrombocytopenia. Its use has been associated with acute tubular
enterohepatic recirculation, with the bulk excreted as a deacylated necrosis.
metabolite in feces and a small amount excreted in the urine.
Dosage adjustment for renal or hepatic insufficiency is not neces-
sary. Usual doses result in serum levels of 5–7 mcg/mL. Rifampin ETHAMBUTOL
is distributed widely in body fluids and tissues. The drug is
relatively highly protein-bound, and adequate cerebrospinal fluid Ethambutol is a synthetic, water-soluble, heat-stable compound,
concentrations are achieved only in the presence of meningeal the dextro-isomer of the structure shown below, dispensed as the
inflammation. dihydrochloride salt.
Rifampin strongly induces most cytochrome P450 isoforms
(CYP1A2, 2C9, 2C19, 2D6, and 3A4), which increases the elimi- CH2OH C2H5
nation of numerous other drugs including methadone, antico- H C NH (CH2)2 NH C H
agulants, cyclosporine, some anticonvulsants, protease inhibitors,
some nonnucleoside reverse transcriptase inhibitors or integrase C2H5 CH2OH

strand transfer inhibitors, contraceptives, and a host of others Ethambutol


846    SECTION VIII  Chemotherapeutic Drugs

Mechanism of Action & Clinical Uses O


N
C NH2
Ethambutol inhibits mycobacterial arabinosyl transferases, which
are encoded by the embCAB operon. Arabinosyl transferases are
involved in the polymerization reaction of arabinoglycan, an
N
essential component of the mycobacterial cell wall. Resistance to
Pyrazinamide (PZA)
ethambutol is due to mutations resulting in overexpression of emb
gene products or within the embB structural gene. Susceptible
strains of Mycobacterium tuberculosis and other mycobacteria are Mechanism of Action & Clinical Uses
inhibited in vitro by ethambutol, 1–5 mcg/mL. Pyrazinamide is converted to pyrazinoic acid—the active form of
Ethambutol is well absorbed from the gut. After ingestion the drug—by mycobacterial pyrazinamidase, which is encoded
of 25 mg/kg, a blood level peak of 2–5 mcg/mL is reached in by pncA. Pyrazinoic acid disrupts mycobacterial cell membrane
2–4 hours. About 20% of the drug is excreted in feces and 50% in metabolism and transport functions. Resistance may be due
urine in unchanged form. Ethambutol accumulates in renal failure, to impaired uptake of pyrazinamide or mutations in pncA that
and the dose should be reduced to three times weekly if creatinine impair conversion of PZA to its active form.
clearance is less than 30 mL/min. Ethambutol crosses the blood- Serum concentrations of 30–50 mcg/mL at 1–2 hours after
brain barrier only when the meninges are inflamed. Concentrations oral administration are achieved with dosages of 25 mg/kg/d.
in cerebrospinal fluid are highly variable, ranging from 4% to 64% Pyrazinamide is well absorbed from the gastrointestinal tract and
of serum levels in the setting of meningeal inflammation. widely distributed in body tissues, including inflamed meninges.
As with all antituberculous drugs, resistance to ethambutol The half-life is 8–11 hours. The parent compound is metabolized
emerges rapidly when the drug is used alone. Therefore, etham- by the liver, but metabolites are renally cleared; therefore, PZA
butol is always given in combination with other antituberculous should be administered at 25–35 mg/kg three times weekly (not
drugs. Ethambutol hydrochloride, 15–25 mg/kg, is usually given daily) in hemodialysis patients and those in whom the creatinine
as a single daily dose in combination with isoniazid, rifampin, and clearance is less than 30 mL/min. In patients with normal renal
pyrazinamide during the initial intensive phase of active tuber- function, a dose of 30–50 mg/kg is used for thrice-weekly or
culosis treatment. The higher dose may be used for treatment of twice-weekly treatment regimens.
tuberculous meningitis. Higher doses have been used with inter- Pyrazinamide is an important front-line drug used in conjunc-
mittent dosing regimens for directly observed therapy; for example, tion with isoniazid and rifampin in short-course (ie, 6-month)
25–30 mg/kg three times weekly or 50 mg/kg administered twice regimens as a “sterilizing” agent active against residual intracellular
weekly. Ethambutol is also used in combination with other agents organisms that may cause relapse. Tubercle bacilli develop resis-
for the treatment of nontuberculous mycobacterial infections, such tance to pyrazinamide fairly readily, but there is no cross-resistance
as Mycobacterium avium complex (MAC) or M. kansasii; the typical with isoniazid or other antimycobacterial drugs.
dose for these infections is 15 mg/kg once daily.
Adverse Reactions
Adverse Reactions Major adverse effects of PZA include hepatotoxicity (in 1–5%
Hypersensitivity to ethambutol is rare. The most common serious of patients), nausea, vomiting, drug fever, photosensitivity, and
adverse event is retrobulbar neuritis, resulting in loss of visual acu- hyperuricemia. The latter occurs uniformly and is not a reason to
ity and red-green color blindness. This dose-related adverse effect halt therapy if patients are asymptomatic.
is more likely to occur at dosages of 25 mg/kg/d continued for
several months. At 15 mg/kg/d or less, visual disturbances occur in
approximately 2% of patients, typically after at least one month of SECOND-LINE DRUGS FOR
treatment. Experts recommend baseline and monthly visual acu- TUBERCULOSIS
ity and color discrimination testing, with particular attention to
patients on higher doses or with impaired renal function. Etham- The alternative drugs listed below are usually considered only (1)
butol is relatively contraindicated in children too young to permit in case of resistance to first-line agents; (2) in case of failure of
assessment of visual acuity and red-green color discrimination. clinical response to conventional therapy; and (3) in case of serious
treatment-limiting adverse drug reactions. Expert guidance is desir-
able in dealing with the toxic effects of these second-line drugs. For
PYRAZINAMIDE many drugs listed in the following text, the dosage, emergence of
resistance, and long-term toxicity have not been fully established.
Pyrazinamide (PZA) is a relative of nicotinamide, and it is used
only for treatment of tuberculosis. It is stable and slightly soluble in
water. It is inactive at neutral pH, but at pH 5.5 it inhibits tubercle Streptomycin
bacilli at concentrations of approximately 20 mcg/mL. The drug is The mechanism of action and other pharmacologic features of
taken up by macrophages and exerts its activity against mycobacte- streptomycin, an aminoglycoside, are discussed in Chapter 45.
ria residing within the acidic environment of lysosomes. The typical adult dosage is 1 g/d (15 mg/kg/d). If the creatinine
CHAPTER 47  Antimycobacterial Drugs    847

clearance is less than 30 mL/min or the patient is on hemodialy- Ethionamide is administered at an initial dose of 250 mg
sis, the dosage is 15 mg/kg two or three times per week. Most once daily, which is increased in 250 mg increments to the
tubercle bacilli are inhibited by streptomycin, 1–10 mcg/mL, recommended dosage of 1 g/d (or 15 mg/kg/d), if possible. The
in vitro. Nontuberculous species of mycobacteria other than 1-g/d dosage, though theoretically desirable, is poorly tolerated
Mycobacterium avium complex (MAC) and Mycobacterium because of gastric irritation and neurologic symptoms, often
kansasii are resistant. All large populations of tubercle bacilli limiting the tolerable daily dose to 500–750 mg. Ethionamide
contain some streptomycin-resistant mutants. On average, 1 is also hepatotoxic. Neurologic symptoms may be alleviated by
in 108 tubercle bacilli can be expected to be resistant to strep- pyridoxine.
tomycin at levels of 10–100 mcg/mL. Resistance may be due Resistance to ethionamide as a single agent develops rapidly in
to a point mutation in either the rpsL gene encoding the S12 vitro and in vivo. There can be low-level cross-resistance between
ribosomal protein or the rrs gene encoding 16S ribosomal RNA, isoniazid and ethionamide.
which alters the ribosomal binding site.
Streptomycin penetrates into cells poorly and is active mainly Capreomycin
against extracellular tubercle bacilli. The drug crosses the blood-
Capreomycin is a peptide protein synthesis inhibitor antibiotic
brain barrier and achieves therapeutic concentrations with
obtained from Streptomyces capreolus. Daily injection of 15 mg/kg
inflamed meninges.
intramuscularly results in peak serum levels of 35–45 mcg/mL
2 hours after a dose. Such concentrations in vitro are inhibitory
Clinical Use in Tuberculosis for many mycobacteria, including multidrug-resistant strains of
Streptomycin sulfate is used when an injectable drug is needed M tuberculosis.
or desirable and in the treatment of infections resistant to other Capreomycin (15 mg/kg/d) is an important injectable agent
drugs. The usual dosage is 15 mg/kg/d intramuscularly or intrave- for treatment of drug-resistant tuberculosis. Strains of M tuber-
nously daily for adults (20–40 mg/kg/d for children, not to exceed culosis that are resistant to streptomycin usually are susceptible
1 g) for several weeks, followed by 15 mg/kg two or three times to capreomycin, though some data suggest cross-resistance with
weekly for several months. Serum concentrations of approxi- strains resistant to amikacin and kanamycin. Resistance to cap-
mately 40 mcg/mL are achieved 30–60 minutes after intramuscu- reomycin, when it occurs, has been associated with rrs, eis, or tlyA
lar injection of a 15 mg/kg dose. Other drugs are always given in gene mutations.
combination to prevent emergence of resistance. Capreomycin is nephrotoxic and ototoxic. Tinnitus, deafness,
and vestibular disturbances occur. The injection causes significant
Adverse Reactions local pain, and sterile abscesses may develop.
Typical dosing of capreomycin is 15 mg/kg/day initially,
Streptomycin is ototoxic and nephrotoxic. Vertigo and hearing which is then reduced to two or three times weekly after an initial
loss are the most common adverse effects and may be permanent. response has been achieved with a daily dosing schedule. The
Toxicity is dose-related, and the risk is increased in the elderly. As intermittent dosing regimen may minimize risk of toxicity.
with all aminoglycosides, the dose must be adjusted according to
renal function (see Chapter 45). Toxicity can be reduced by limit-
ing therapy to no more than 6 months whenever possible.
Cycloserine
Cycloserine—a structural analog of d-alanine—inhibits cell
wall synthesis, as discussed in Chapter 43. Concentrations of
Ethionamide
15–20 mcg/mL inhibit many strains of M tuberculosis. The usual
Ethionamide is chemically related to isoniazid and similarly blocks dosage of cycloserine in tuberculosis is 0.5–1 g/d in two divided
the synthesis of mycolic acids. It is poorly water soluble and avail- oral doses. The drug is widely distributed to tissues, including
able only for oral use. It is metabolized by the liver. the central nervous system. This drug is cleared renally, and the
N dose should be reduced by half if creatinine clearance is less than
H5C2
50 mL/min. Alternatively, it may be reduced to 500 mg three
times weekly.
The most serious toxic effects are peripheral neuropathy and
C central nervous system dysfunction, including depression and
S NH2 psychoses. Pyridoxine, 100 mg or more per day, should be given
Ethionamide with cycloserine because this ameliorates neurologic toxicity.
Adverse effects, which are most common during the first 2 weeks
Most tubercle bacilli are inhibited in vitro by ethionamide, of therapy, occur in 25% or more of patients, especially at higher
2.5 mcg/mL or less. Some other species of mycobacteria also are doses leading to peak concentrations greater than 35 mcg/mL.
inhibited by ethionamide, 10 mcg/mL. Serum concentrations in Adverse effects can be minimized by monitoring peak serum
plasma and tissues of approximately 1-5 mcg/mL are achieved by concentrations. The peak concentration is reached 2–4 hours
a dosage of 1 g/d. Cerebrospinal fluid concentrations are equal to after dosing. The recommended range of peak concentrations is
those in serum. 20–35 mcg/mL.
848    SECTION VIII  Chemotherapeutic Drugs

Aminosalicylic Acid (PAS) between streptomycin and amikacin, but kanamycin resistance
often indicates resistance to amikacin as well. Peak serum con-
Aminosalicylic acid is a folate synthesis antagonist that is active
centrations of 30–45 mcg/mL are achieved 30–60 minutes after
almost exclusively against M tuberculosis. It is structurally similar
a 15-mg/kg intravenous infusion or intramuscular injection.
to p-amino-benzoic acid (PABA) and is thought to have a similar
Amikacin is indicated for treatment of tuberculosis suspected
mechanism of action to the sulfonamides (see Chapter 46). In the
or known to be caused by streptomycin-resistant or multidrug-
USA, PAS is commercially available as a 4-g packet of delayed-
resistant strains. This drug must be used in combination with at
release granules. In order to protect the integrity of the delayed-
least one and preferably two or three other drugs to which the
release coating, the granules must be administered sprinkled over
isolate is susceptible for treatment of drug-resistant cases. The
applesauce or yogurt, or swirled in fruit juice and swallowed
recommended dosage is 15 mg/kg once daily initially, followed by
whole.
intermittent dosing two or three times per week.
COOH

OH Fluoroquinolones
In addition to their activity against many Gram-positive and
Gram-negative bacteria (discussed in Chapter 46), ciprofloxacin,
levofloxacin, gatifloxacin, and moxifloxacin inhibit strains of
NH2
M tuberculosis at concentrations less than 2 mcg/mL. They are
Aminosalicylic acid (PAS) also active against atypical mycobacteria. Moxifloxacin is the
most active against M tuberculosis in vitro. Levofloxacin tends to
Tubercle bacilli are usually inhibited in vitro by aminosalicylic be slightly more active than ciprofloxacin against M tuberculo-
acid, 1–5 mcg/mL. The granule formulation of aminosalicylic sis, whereas ciprofloxacin is slightly more active against atypical
acid results in improved absorption from the gastrointestinal mycobacteria.
tract. Peak serum levels are expected to be 20–60 mcg/mL 6 hours Fluoroquinolones are an important addition to the drugs avail-
after a 4 g oral dose. The dosage is 8–12 g/d orally for adults and able for tuberculosis, especially for strains that are resistant to first-
300 mg/kg/d for children, administered in two or three divided line agents. The World Health Organization recommends using a
doses. The drug is widely distributed in tissues and body fluids later generation fluoroquinolone such as moxifloxacin or levoflox-
except the cerebrospinal fluid. Aminosalicylic acid is rapidly acin. Resistance, which may result from one of several single point
excreted in the urine, in part as active PAS and in part as the mutations in the gyrase A subunit, develops rapidly if a fluoroqui-
acetylated compound and other metabolic products. To avoid nolone is used as a single agent; thus, the drug must be used in
accumulation in renal impairment, the maximum dose is 4 g twice combination with two or more additional active agents. Typically,
daily when creatinine clearance is less than 30 mL/min. Very high resistance to one fluoroquinolone indicates class resistance. How-
concentrations of aminosalicylic acid are reached in the urine, ever, moxifloxacin may retain some activity in strains resistant to
which can result in crystalluria. ofloxacin. The dosage of levofloxacin is 500–750 mg once a day,
Aminosalicylic acid is used infrequently in the USA because and some clinicians increase to 1000 mg daily if tolerated. The
other oral drugs are better tolerated. Gastrointestinal symptoms dosage of moxifloxacin is 400 mg once a day. Some experts rec-
are common but occur less frequently with the delayed-release ommend checking peak serum concentrations. Expected levels at
granules; they may be diminished by giving the drug with meals about two hours post-dose are 8–12 mcg/mL for levofloxacin and
and with antacids. Peptic ulceration and hemorrhage may occur. 3–5 mcg/mL for moxifloxacin.
Hypersensitivity reactions manifested by fever, joint pains, skin
rashes, hepatosplenomegaly, hepatitis, adenopathy, and granulo- Linezolid
cytopenia often occur after 3–8 weeks of PAS therapy, making it
necessary to stop administration temporarily or permanently. Linezolid (discussed in Chapter 44) inhibits strains of M tuberculosis
in vitro at concentrations of 4–8 mcg/mL. It achieves good intra-
cellular concentrations, and it is active in murine models of
Kanamycin & Amikacin tuberculosis. Linezolid has been used in combination with other
The aminoglycoside antibiotics are discussed in Chapter 45. second- and third-line drugs to treat patients with tuberculosis
Kanamycin had been used for treatment of tuberculosis caused by caused by multidrug-resistant strains. Conversion of sputum
streptomycin-resistant strains, but it is no longer available in the cultures to negative was associated with linezolid use in these
USA, and less toxic alternatives (eg, capreomycin and amikacin) cases. Significant adverse effects, including bone marrow suppres-
have taken its place. sion and irreversible peripheral and optic neuropathy, have been
Amikacin is playing a greater role in the treatment of tuberculo- reported with the prolonged courses of therapy that are necessary
sis due to the prevalence of multidrug-resistant strains. Prevalence for treatment of tuberculosis. A 600-mg (adult) dose administered
of amikacin-resistant strains is low (<5%), and most multidrug- once a day (half of that used for treatment of other bacterial infec-
resistant strains remain amikacin-susceptible. M tuberculosis is tions) seems to be sufficient and may limit the occurrence of these
inhibited at concentrations of 1 mcg/mL or less. Amikacin is also adverse effects. Experts recommend supplemental pyridoxine for
active against atypical mycobacteria. There is no cross-resistance patients treated with linezolid. Although linezolid may prove to
CHAPTER 47  Antimycobacterial Drugs    849

be an important new agent for treatment of tuberculosis, at this with HIV infection because of an unacceptably high relapse rate
point it should be used only for multidrug-resistant strains that with rifampin-resistant organisms. Rifapentine in combination
also are resistant to several other first- and second-line agents. It is with isoniazid, typically both dosed at 900 mg once weekly for
generally avoided in patients on concomitant serotonergic agents 3 months (12 doses each in total), is an effective short course treat-
due to concern for serotonin syndrome. ment for latent tuberculosis infection.

Rifabutin Bedaquiline
Rifabutin is derived from rifamycin and is related to rifampin. Bedaquiline, a diarylquinoline, is the first drug with a novel mech-
It has significant activity against M tuberculosis, MAC, and anism of action against M tuberculosis to be approved since 1971.
Mycobacterium fortuitum (see below). Its activity is similar to Bedaquiline inhibits adenosine 5′-triphosphate (ATP) synthase
that of rifampin, and cross-resistance with rifampin is virtually in mycobacteria, has in vitro activity against both replicating and
complete. Some rifampin-resistant strains may appear susceptible nonreplicating bacilli, and has bactericidal and sterilizing activity
to rifabutin in vitro, but a clinical response is unlikely because in the murine model of tuberculosis. Cross-resistance has been
the molecular basis of resistance, rpoB mutation, is the same. reported between bedaquiline and clofazimine, likely via upregu-
Rifabutin is both substrate and inducer of cytochrome P450 lation of the multisubstrate efflux pump, MmpL5.
enzymes. Because it is a less potent inducer, rifabutin is often Peak plasma concentration and plasma exposure to bedaquiline
used in place of rifampin for treatment of tuberculosis in patients increase approximately twofold when administered with high-fat
with HIV infection who are receiving antiretroviral therapy with food. Bedaquiline is highly protein-bound (>99%), is metabolized
a protease inhibitor, a nonnucleoside reverse transcriptase inhibi- chiefly through the cytochrome P450 system, and is excreted
tor (eg, efavirenz), or an integrase strand transfer inhibitor (eg primarily via the feces. The mean terminal half-life of bedaquiline
dolutegravir), drugs that also are cytochrome P450 or UDP gluc- and its major metabolite (M2), which is four to six times less
uronosyltransferase (UGT) substrates. active in terms of antimycobacterial potency, is approximately
The typical dosage of rifabutin is 300 mg/d unless the patient 5.5 months. This long elimination phase probably reflects slow
is receiving a protease inhibitor, in which case the dosage should release of bedaquiline and M2 from peripheral tissues. CYP3A4 is
be reduced, typically by half. If efavirenz (also a cytochrome the major isoenzyme involved in the metabolism of bedaquiline,
P450 inducer) is used, the recommended dosage of rifabutin is and potent inhibitors or inducers of this enzyme cause clinically
600 mg/d. Rifabutin may accumulate in severe renal impairment, significant drug interactions.
and the dose should be reduced by half if creatinine clearance is Current recommendations state that bedaquiline, in combina-
less than 30 mL/min. Rifabutin is associated with similar rates tion with at least three other active medications, may be used for
of hepatotoxicity or rash compared to rifampin; it can also cause 24 weeks of treatment in adults with laboratory-confirmed pul-
leukopenia, thrombocytopenia, and optic neuritis. monary tuberculosis if the isolate is resistant to both isoniazid and
rifampin. The recommended dosage for bedaquiline is 400 mg
Rifapentine once daily orally for 2 weeks, followed by 200 mg three times a
week for 22 weeks taken orally with food in order to maximize
Rifapentine is another analog of rifampin. It is active against both absorption. The most common adverse effects, occurring at rates
M tuberculosis and MAC. As with all rifamycins, it is a bacterial of 25% or more, are nausea, arthralgia, and headache. Bedaquiline
RNA polymerase inhibitor, and cross-resistance between rifampin has been associated with both hepatotoxicity and cardiac toxicity.
and rifapentine is complete. Like rifampin, rifapentine is a potent The FDA has issued a black-box warning related to the risk of
inducer of cytochrome P450 enzymes, and it has the same drug QTc prolongation and associated mortality. It should be reserved
interaction profile; however, when rifapentine is administered for patients who do not have other treatment options and used
intermittently, induction of metabolism of other medications is with caution in patients with other risk factors for cardiac conduc-
less pronounced compared to rifampin. Toxicity is similar to that tion abnormalities.
of rifampin. Rifapentine and its microbiologically active metabo-
lite, 25-desacetylrifapentine, have an elimination half-life of
13 hours. Rifapentine, 600 mg (10 mg/kg) once or twice weekly, ■■ DRUGS ACTIVE AGAINST
has been used for treatment of tuberculosis caused by rifampin-
susceptible strains during the continuation phase (ie, after the NONTUBERCULOUS
first 2 months of therapy and ideally after conversion of sputum MYCOBACTERIA
cultures to negative); however, this regimen has decreased efficacy
compared with the standard rifampin-based regimen. Revised Many mycobacterial infections seen in clinical practice in the
guidelines for treatment of drug-susceptible tuberculosis pub- United States are caused by nontuberculous mycobacteria (NTM),
lished in 2016 recommend against it. In particular, its use should formerly known as “atypical mycobacteria.” These organisms have
be avoided in patients at higher risk of failure, including those distinctive laboratory characteristics, are present in the environ-
with positive cultures at the end of the intensive treatment phase ment, and are generally not communicable from person to person.
and those with evidence of cavitation on chest radiographs. Rifa- As a rule, these mycobacterial species are less susceptible than
pentine should not be used to treat active tuberculosis in patients M tuberculosis to antituberculous drugs. On the other hand, agents
850    SECTION VIII  Chemotherapeutic Drugs

TABLE 47–3  Clinical features and treatment options for infections with atypical mycobacteria.
Species Clinical Features Treatment Options

M kansasii Resembles tuberculosis Amikacin, clarithromycin, ethambutol, isoniazid, moxifloxacin, rifampin,


streptomycin, trimethoprim-sulfamethoxazole
M marinum Granulomatous cutaneous disease Amikacin, clarithromycin, ethambutol, doxycycline, levofloxacin, minocycline,
rifampin, trimethoprim-sulfamethoxazole
M scrofulaceum Cervical adenitis in children Amikacin, erythromycin (or other macrolide), rifampin, streptomycin
(Surgical excision is often curative and the treatment of choice.)
M avium complex Pulmonary disease in patients with chronic Amikacin, azithromycin, clarithromycin, ethambutol, moxifloxacin, rifabutin
(MAC) lung disease; disseminated infection in AIDS
M chelonae Abscess, sinus tract, ulcer; bone, joint, tendon Amikacin, doxycycline, imipenem, linezolid, macrolides, tobramycin
infection
M fortuitum Abscess, sinus tract, ulcer; bone, joint, tendon Amikacin, cefoxitin, ciprofloxacin, doxycycline, imipenem, minocycline,
infection moxifloxacin, ofloxacin, trimethoprim-sulfamethoxazole
M ulcerans Skin ulcers Clarithromycin, isoniazid, streptomycin, rifampin, minocycline, moxifloxacin
(Surgical excision may be effective.)

such as macrolides, sulfonamides, and tetracyclines, which are not DAPSONE & OTHER SULFONES
active against M tuberculosis, may be effective for infections caused
by NTM. Emergence of resistance during therapy is also a prob- Several drugs closely related to the sulfonamides have been used
lem with these mycobacterial species, and active infection should effectively in the long-term treatment of leprosy. The most widely
be treated with combinations of drugs. M kansasii is susceptible used is dapsone (diaminodiphenylsulfone). Like the sulfon-
to rifampin and ethambutol, partially susceptible to isoniazid, and amides, it inhibits folate synthesis. Resistance can emerge in large
completely resistant to pyrazinamide. A three-drug combination populations of M leprae, eg, in lepromatous leprosy, particularly
of isoniazid, rifampin, and ethambutol is the conventional treat- if low doses are given. Therefore, the combination of dapsone,
ment for M kansasii infection. A few representative pathogens, rifampin, and clofazimine is recommended for initial therapy of
with the clinical presentation and the drugs to which they are lepromatous leprosy. A combination of dapsone plus rifampin is
often susceptible, are given in Table 47–3. commonly used for leprosy with a lower organism burden. Dap-
M avium complex (MAC), which includes both M avium sone may also be used to prevent and treat Pneumocystis jiroveci
and M intracellulare, is an important and common cause of dis- pneumonia in AIDS patients.
seminated disease in late stages of AIDS (CD4 counts < 50/μL).
O
MAC is much less susceptible than M tuberculosis to most anti-
tuberculous drugs. Combinations of agents are required to sup- NH2 S NH2
press the infection. Azithromycin, 500–600 mg once daily, or O
clarithromycin, 500 mg twice daily, plus ethambutol, 15 mg/kg/d, Dapsone
is an effective and well-tolerated regimen for treatment of dissemi-
nated disease. Some authorities recommend use of a third agent, Sulfones are well absorbed from the gut and widely distributed
especially rifabutin, 300 mg once daily. Other agents that may be throughout body fluids and tissues. Dapsone’s half-life is 1–2 days,
useful are listed in Table 47–3. Azithromycin and clarithromycin and drug tends to be retained in skin, muscle, liver, and kidney. Skin
are the prophylactic drugs of choice for preventing disseminated heavily infected with M leprae may contain several times more drug
MAC in AIDS patients with CD4 cell counts less than 50/μL. than normal skin. Sulfones are excreted into bile and reabsorbed
Rifabutin in a single daily dose of 300 mg has been shown to in the intestine. Excretion into urine is variable, and most excreted
reduce the incidence of MAC bacteremia but is less effective than drug is acetylated. In renal failure, the dose may have to be adjusted.
macrolides. The usual adult dosage in leprosy is 100 mg daily. For children, the
dose is proportionately less, depending on weight.
Dapsone is usually well tolerated. Many patients develop some
■■ DRUGS USED IN LEPROSY hemolysis, particularly if they have glucose-6-phosphate dehydro-
genase deficiency. Methemoglobinemia is common but usually
Mycobacterium leprae has never been grown in vitro, but animal is not clinically significant. Gastrointestinal intolerance, fever,
models, such as growth in injected mouse footpads, have permit- pruritus, and rash occur. During dapsone therapy of lepromatous
ted laboratory evaluation of drugs. Only those drugs with the wid- leprosy, erythema nodosum leprosum often develops. It is some-
est clinical use are presented here. Because of increasing reports of times difficult to distinguish reactions to dapsone from manifesta-
dapsone resistance, treatment of leprosy with combinations of the tions of the underlying illness. Erythema nodosum leprosum may
drugs listed below is recommended. be suppressed by thalidomide (see Chapter 55).
CHAPTER 47  Antimycobacterial Drugs    851

RIFAMPIN and a major portion of the drug is excreted in feces. Clofazimine


is stored widely in reticuloendothelial tissues and skin, and its
Rifampin (see earlier discussion) in a dosage of 600 mg daily is crystals can be seen inside phagocytic reticuloendothelial cells. It
highly effective in leprosy and is given with at least one other drug is slowly released from these deposits, so the serum half-life may
to prevent emergence of resistance. Even a dose of 600 mg per be 2 months. A common dosage of clofazimine is 100–200 mg/d
month may be beneficial in combination therapy. orally. The most prominent adverse effect is discoloration of the
skin and conjunctivae. Gastrointestinal side effects are common.
This medication is no longer commercially available, but it can
CLOFAZIMINE be obtained through established programs. For example, an inves-
tigational new drug (IND) program is established in the USA
Clofazimine is a phenazine dye used in the treatment of multi- through the National Hansen’s Disease Program. Internation-
bacillary leprosy, which is defined as having a positive smear from ally, ministries of health can make requests directly to the World
any site of infection. Its mechanism of action has not been clearly Health Organization.
established. Absorption of clofazimine from the gut is variable,

SUMMARY  First-Line Antimycobacterial Drugs


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

ISONIAZID Inhibits synthesis of mycolic Bactericidal activity against First-line agent for Oral, IV • hepatic clearance (half-life 1 h)
acids, an essential susceptible strains of tuberculosis • treatment of • reduces levels of phenytoin • Toxicity:
component of mycobacterial M tuberculosis latent infection • less active Hepatotoxic, peripheral neuropathy (give
cell walls against nontuberculous pyridoxine to prevent)
mycobacteria

RIFAMYCINS

  •  Rifampin Inhibits DNA-dependent RNA Bactericidal activity against First-line agent for Oral, IV • hepatic clearance (half-life 3.5 h)
polymerase, thereby blocking susceptible bacteria and tuberculosis • nontuberculous • potent cytochrome P450 inducer • turns
production of RNA mycobacteria • resistance mycobacterial infections body fluids orange color • Toxicity: Rash,
rapidly emerges when used • eradication of nephritis, thrombocytopenia, cholestasis,
as a single drug in the meningococcal colonization, flu-like syndrome with intermittent
treatment of active infection staphylococcal infections dosing

  •  Rifabutin: Oral; similar to rifampin but less cytochrome P450 induction and fewer drug interactions
  • Rifapentine: Oral; long-acting analog of rifampin that may be given once weekly in select cases during the continuation phase of tuberculosis treatment or for treatment
of latent tuberculosis

PYRAZINAMIDE Not fully understood • Bacteriostatic activity against “Sterilizing” agent used Oral • hepatic clearance (half-life 9 h), but
pyrazinamide is converted to susceptible strains of during first 2 months of metabolites are renally cleared so use
the active pyrazinoic acid M tuberculosis • may be therapy • allows total 3 doses weekly if creatinine clearance
under acidic conditions in bactericidal against actively duration of therapy to be <30 mL/min • Toxicity: Hepatotoxic,
macrophage lysosomes dividing organisms shortened to 6 months hyperuricemia

ETHAMBUTOL Inhibits mycobacterial Bacteriostatic activity against Given in four-drug initial Oral • mixed clearance (half-life 4 h)
arabinosyl transferases, which susceptible mycobacteria combination therapy for • dose must be reduced in renal failure
are involved in the tuberculosis until drug • Toxicity: Retrobulbar neuritis
polymerization reaction of sensitivities are known • also
arabinoglycan, an essential used for nontuberculous
component of the mycobacterial infections
mycobacterial cell wall
852    SECTION VIII  Chemotherapeutic Drugs

P R E P A R A T I *O N S Centers for Disease Control and Prevention (CDC): Update: Adverse event data
and revised American Thoracic Society/CDC recommendations against
A V A I L A B L E the use of rifampin and pyrazinamide for treatment of latent tubercu-
losis infection—United States, 2003. MMWR Morb Mortal Wkly Rep
2003;52:735.
GENERIC NAME AVAILABLE AS
Curry International Tuberculosis Center and California Department of Public
DRUGS USED IN TUBERCULOSIS Health, 2016: Drug-Resistant Tuberculosis: A Survival Guide for Clinicians,
Aminosalicylic acid Paser Third Edition [1-305].
Bedaquiline fumarate Sirturo Gillespie SH et al: Early bactericidal activity of a moxifloxacin and isoniazid
combination in smear-positive pulmonary tuberculosis. J Antimicrob
Capreomycin Capastat Chemother 2005;56:1169.
Ethambutol Generic, Myambutol Griffith DE et al: An official ATS/IDSA statement: Diagnosis, treatment, and
Ethionamide Trecator, Trecator-SC prevention of nontuberculous mycobacterial disease. Am J Respir Crit Care
Isoniazid Generic Med 2007;175:367.
Hugonnet J-E et al: Meropenem-clavulanate is effective against extensively drug-
Pyrazinamide Generic
resistant Mycobacterium tuberculosis. Science 2009;323:1215.
Rifabutin Generic, Mycobutin Jasmer RM, Nahid P, Hopewell PC: Latent tuberculosis infection. N Engl J Med
Rifampin Generic, Rifadin, Rimactane 2002;347:1860.
Rifapentine Priftin Kinzig-Schippers M et al: Should we use N-acetyltransferase type 2 genotyping to
Streptomycin Generic personalize isoniazid doses? Antimicrob Agents Chemother 2005;49:1733.
Lee M et al: Linezolid for treatment of chronic extensively drug-resistant tubercu-
DRUGS USED IN LEPROSY
losis. N Engl J Med 2012;367:1508.
Clofazimine Lamprene Mitnick CD et al: Comprehensive treatment of extensively drug-resistant tubercu-
Dapsone Generic losis. N Engl J Med 2008;359:563.
*
Nahid P et al: Official American Thoracic Society/Centers for Disease Control
Drugs used against nontuberculous mycobacteria are listed in Chapters 43–46. and Prevention/Infectious Diseases Society of America Clinical Practice
Guidelines: Treatment of Drug-Susceptible Tuberculosis. Clin Infect Dis
REFERENCES 2016;63:e147.
Sulochana S, Rahman F, Paramasivan CN: In vitro activity of fluoroquinolones
Centers for Disease Control and Prevention (CDC): Provisional CDC Guidelines against Mycobacterium tuberculosis. J Chemother 2005;17:169.
for the use and safety monitoring of bedaquiline fumarate (Sirturo) for the
treatment of multidrug-resistant tuberculosis. MMWR Morb Mortal Wkly Targeted tuberculin testing and treatment of latent tuberculosis infection. Am J
Rep 2013;62:1. Respir Crit Care Med 2000;161(4 Part 2):S221.
Centers for Disease Control and Prevention (CDC): Recommendations for use Zhang Y, Yew WW: Mechanisms of drug resistance in Mycobacterium tuberculosis.
of an isoniazid-rifapentine regimen with direct observation to treat latent Int J Tuberc Lung Dis 2009;13:1320.
Mycobacterium tuberculosis infection. MMWR Morb Mortal Wkly Rep Zumla A et al: Current concepts—Tuberculosis. N Engl J Med 2013;368:745.
2011;60:1650.

C ASE STUDY ANSWER

The patient should be started on four-drug therapy with If dolutegravir is chosen, it must be administered twice daily
rifampin, isoniazid, pyrazinamide, and ethambutol. He should due to the interaction with rifampin; alternatively, rifabutin
also be started on antiretroviral therapy for HIV. If a protease- can be used in place of rifampin, and dolutegravir can be
inhibitor-based antiretroviral regimen is used to treat his HIV, dosed once daily. The patient is at increased risk of developing
rifabutin should replace rifampin because of the serious drug- hepatotoxicity from both isoniazid and pyrazinamide given
drug interactions between rifampin and protease inhibitors. his history of alcohol use.
51
C H A P T E R

Clinical Use of
Antimicrobial Agents
Harry W. Lampiris, MD, &
Daniel S. Maddix, PharmD

C ASE STUDY

A 65-year-old man undergoes cystoscopy because of the 90/50, pulse of 120, temperature of 38.5° C and respira-
presence of microscopic hematuria in order to rule out tory rate of 24. The patient is disoriented but the physical
urologic malignancy. The patient has mild dysuria and exam is otherwise unremarkable. Laboratory test shows
pyuria and empirically receives oral therapy with cip- WBC 24,000/mm3 and elevated serum lactate; urinalysis
rofloxacin for presumed urinary tract infection prior to shows 300 WBC per high power field and 4+ bacteria.
the procedure and tolerates the procedure well. Approxi- What possible organisms are likely to be responsible for
mately 48 hours after the procedure, the patient presents the patient’s symptoms? At this point, what antibiotic(s)
to the emergency department with confusion, dysuria would you choose for initial therapy of this potentially
and chills. Physical exam reveals a blood pressure of life-threatening infection?

The development of antimicrobial drugs represents one of the 3. What are the likely etiologic agents for the patient’s illness?
most important advances in therapeutics, both in the control 4. What measures should be taken to protect individuals exposed
or cure of serious infections and in the prevention and treat- to the index case to prevent secondary cases, and what measures
ment of infectious complications of other therapeutic modalities should be implemented to prevent further exposure?
such as cancer chemotherapy, immunosuppression, and surgery. 5. Is there clinical evidence (eg, from well-executed clinical trials) that
However, evidence is overwhelming that antimicrobial agents antimicrobial therapy will confer clinical benefit for the patient?
are vastly overprescribed in outpatient settings in the USA, and
the availability of antimicrobial agents without prescription in Once a specific cause is identified based on specific microbio-
many developing countries has—by facilitating the development logic tests, the following further questions should be considered:
of resistance—already severely limited therapeutic options in the 1. If a specific microbial pathogen is identified, can a narrower-
treatment of life-threatening infections. Therefore, the clinician spectrum agent be substituted for the initial empiric drug?
should first determine whether antimicrobial therapy is warranted 2. Is one agent or a combination of agents necessary?
for a given patient. The specific questions one should ask include
3. What are the optimal dose, route of administration, and dura-
the following:
tion of therapy?
1. Is an antimicrobial agent indicated on the basis of clinical find- 4. What specific tests (eg, susceptibility testing) should be under-
ings? Or is it prudent to wait until such clinical findings taken to identify patients who will not respond to treatment?
become apparent? 5. What adjunctive measures can be undertaken to eradicate
2. Have appropriate clinical specimens been obtained to establish the infection? For example, is surgery feasible for removal
a microbiologic diagnosis? of devitalized tissue or foreign bodies—or drainage of an

904
CHAPTER 51  Clinical Use of Antimicrobial Agents     905

abscess—into which antimicrobial agents may be unable to testing, polymerase chain reaction, and serology) also may confirm
penetrate? Is it possible to decrease the dosage of immunosup- specific etiologic agents.
pressive therapy in patients who have undergone organ trans-
plantation? Is it possible to reduce morbidity or mortality due C. Formulate a Microbiologic Diagnosis
to the infection by reducing host immunologic response to the The history, physical examination, and immediately available lab-
infection (eg, by the use of corticosteroids for the treatment oratory results (eg, Gram stain of urine or sputum) may provide
of severe Pneumocystis jiroveci pneumonia or meningitis due to highly specific information. For example, in a young man with
Streptococcus pneumoniae)? urethritis and a Gram-stained smear from the urethral meatus
demonstrating intracellular Gram-negative diplococci, the most
likely pathogen is Neisseria gonorrhoeae. In the latter instance,
■■ EMPIRIC ANTIMICROBIAL however, the clinician should be aware that a significant number
of patients with gonococcal urethritis have negative Gram stains
THERAPY for the organism and that a significant number of patients with
gonococcal urethritis harbor concurrent chlamydial infection that
Antimicrobial agents are frequently used before the pathogen is not demonstrated on the Gram-stained smear.
responsible for a particular illness or the susceptibility to a par-
ticular antimicrobial agent is known. This use of antimicrobial D. Determine the Necessity for Empiric Therapy
agents is called empiric (or presumptive) therapy and is based on
Whether to initiate empiric therapy is an important clinical deci-
experience with a particular clinical entity. The usual justifica-
sion based partly on experience and partly on data from clinical
tion for empiric therapy is the hope that early intervention will
trials. Empiric therapy is indicated when there is a significant risk
improve the outcome; in the best cases, this has been established
of serious morbidity or mortality if therapy is withheld until a
by placebo-controlled, double-blind, prospective clinical trials.
specific pathogen is detected by the clinical laboratory.
For example, treatment of febrile episodes in neutropenic cancer
In other settings, empiric therapy may be indicated for public
patients with empiric antimicrobial therapy has been demon-
health reasons rather than for demonstrated superior outcome of
strated to have impressive morbidity and mortality benefits even
therapy in a specific patient. For example, urethritis in a young
though the specific bacterial agent responsible for fever is deter-
sexually active man usually requires treatment for N gonorrhoeae
mined for only a minority of such episodes.
and Chlamydia trachomatis despite the absence of microbiologic
Finally, there are many clinical entities, such as certain episodes
confirmation at the time of diagnosis. Because the risk of noncom-
of community-acquired pneumonia, in which it is difficult to
pliance with follow-up visits in this patient population may lead
identify a specific pathogen. In such cases, a clinical response to
empiric therapy may be an important clue to the likely pathogen. to further transmission of these sexually transmitted pathogens,
Frequently, the signs and symptoms of infection diminish as empiric therapy is warranted.
a result of empiric therapy, and microbiologic test results become
available to establish a specific microbiologic diagnosis. At the E. Institute Treatment
time that the pathogenic organism responsible for the illness is Selection of empiric therapy may be based on the microbiologic
identified, empiric therapy is optimally modified to definitive diagnosis or a clinical diagnosis without available microbiologic
therapy, which is typically narrower in coverage and is given for clues. If no microbiologic information is available, the antimicro-
an appropriate duration based on the results of clinical trials, or bial spectrum of the agent or agents chosen must necessarily be
experience when clinical trial data are not available. broader, taking into account the most likely pathogens responsible
for the patient’s illness.

Approach to Empiric Therapy Choice of Antimicrobial Agent


Initiation of empiric therapy should follow a specific and system-
Selection from among several drugs depends on host factors that
atic approach.
include the following: (1) concomitant disease states (eg, AIDS,
neutropenia due to the use of cytotoxic chemotherapy, organ
A. Formulate a Clinical Diagnosis of Microbial Infection transplantation, severe chronic liver or kidney disease) or the use of
Using all available data, the clinician should determine that there immunosuppressive medications; (2) prior adverse drug effects; (3)
is a clinical syndrome compatible with infection (eg, pneumonia, impaired elimination or detoxification of the drug (may be geneti-
cellulitis, sinusitis). cally predetermined but more frequently is associated with impaired
renal or hepatic function due to underlying disease); (4) age of the
B. Obtain Specimens for Laboratory Examination patient; (5) pregnancy status; and (6) epidemiologic exposure (eg,
Examination of stained specimens by microscopy or simple exposure to a sick family member or pet, recent hospitalization,
examination of an uncentrifuged sample of urine for white blood recent travel, occupational exposure, or new sexual partner).
cells and bacteria may provide important immediate etiologic Pharmacologic factors include (1) the kinetics of absorption,
clues. Cultures of selected anatomic sites (blood, sputum, urine, distribution, and elimination; (2) the ability of the drug to be
cerebrospinal fluid, and stool) and nonculture methods (antigen delivered to the site of infection; (3) the potential toxicity of an
906    SECTION VIII  Chemotherapeutic Drugs

agent; and (4) pharmacokinetic or pharmacodynamic interactions Tests measure the concentration of drug required to inhibit
with other drugs. growth of the organism (minimal inhibitory concentration
Knowledge of the susceptibility of an organism to a specific [MIC]) or to kill the organism (minimal bactericidal concen-
agent in a hospital or community setting is important in the tration [MBC]). The results of these tests can then be correlated
selection of empiric therapy. Pharmacokinetic differences among with known drug concentrations in various body compartments.
agents with similar antimicrobial spectrums may be exploited to Only MICs are routinely measured in most infections, whereas
reduce the frequency of dosing (eg, ceftriaxone, ertapenem, or in infections in which bactericidal therapy is required for eradi-
daptomycin may be conveniently given once every 24 hours). cation of infection (eg, meningitis, endocarditis, sepsis in the
Finally, increasing consideration is being given to the cost of granulocytopenic host), MBC measurements occasionally may
antimicrobial therapy, especially when multiple agents with com- be useful.
parable efficacy and toxicity are available for a specific infection.
Changing from intravenous to oral antibiotics for prolonged Specialized Assay Methods
administration can be particularly cost-effective.
A. Beta-Lactamase Assay
Brief guides to empiric therapy based on presumptive microbial
diagnosis and site of infection are given in Tables 51–1 and 51–2. For some bacteria (eg, Haemophilus species), the susceptibility
patterns of strains are similar except for the production of β lac-
tamase. In these cases, extensive susceptibility testing may not be
required, and a direct test for β lactamase using a chromogenic
■■ ANTIMICROBIAL THERAPY β-lactam substrate (nitrocefin disk) may be substituted.
OF INFECTIONS WITH KNOWN
ETIOLOGY B. Synergy Studies
Synergy studies are in vitro tests that attempt to measure syner-
INTERPRETATION OF CULTURE RESULTS gistic, additive, indifferent, or antagonistic drug interactions. In
general, these tests have not been standardized and have not cor-
Properly obtained and processed specimens for culture frequently related well with clinical outcome. (See section on Antimicrobial
yield reliable information about the cause of infection. The lack of a Drug Combinations for details.)
confirmatory microbiologic diagnosis may be due to the following:
1. Sample error, eg, obtaining cultures after antimicrobial agents MONITORING THERAPEUTIC
have been administered, inadequate volume or quantity of speci- RESPONSE: DURATION OF THERAPY
men obtained, or contamination of specimens sent for culture
2. Noncultivable or slow-growing organisms (Histoplasma capsu- The therapeutic response may be monitored microbiologically or
latum, Bartonella or Brucella species), in which cultures are clinically. Cultures of specimens taken from infected sites should
often discarded before sufficient growth has occurred for eventually become sterile or demonstrate eradication of the patho-
detection gen and are useful for documenting recurrence or relapse. Follow-
3. Requesting bacterial cultures when infection is due to other up cultures may also be useful for detecting superinfections or the
organisms development of resistance. Clinically, the patient’s systemic mani-
4. Not recognizing the need for special media or isolation tech- festations of infection (malaise, fever, leukocytosis) should abate,
niques (eg, charcoal yeast extract agar for isolation of Legionella and the clinical findings should improve (eg, as shown by clearing
species, shell-vial tissue culture system for rapid isolation of of radiographic infiltrates or lessening hypoxemia in pneumonia).
cytomegalovirus) The duration of definitive therapy required for cure depends on
the pathogen, the site of infection, and host factors (immunocom-
Even in the setting of a classic infectious disease for which promised patients generally require longer courses of treatment).
isolation techniques have been established for decades (eg, pneu- Precise data on duration of therapy exist for some infections (eg,
mococcal pneumonia, pulmonary tuberculosis, streptococcal streptococcal pharyngitis, syphilis, gonorrhea, tuberculosis, and
pharyngitis), the sensitivity of the culture technique may be inad- cryptococcal meningitis). In many other situations, duration of
equate to identify all cases of the disease. therapy is determined empirically. Minimizing duration of anti-
microbial therapy for specific infections is an intervention that
may help prevent the development of antimicrobial resistance.
GUIDING ANTIMICROBIAL THERAPY OF For many infections, a combined medical-surgical approach may
ESTABLISHED INFECTIONS be required for clinical cure.

Susceptibility Testing Clinical Failure of Antimicrobial Therapy


Testing bacterial pathogens in vitro for their susceptibility to When the patient has an inadequate clinical or microbiologic
antimicrobial agents is extremely valuable in confirming suscepti- response to antimicrobial therapy selected by in vitro suscepti-
bility, ideally to a narrow-spectrum nontoxic antimicrobial drug. bility testing, systematic investigation should be undertaken to
CHAPTER 51  Clinical Use of Antimicrobial Agents     907

TABLE 51–1  Empiric antimicrobial therapy based on microbiologic etiology.


Suspected or Proven Disease or Pathogen Drugs of First Choice Alternative Drugs

Gram-negative cocci (aerobic)


  Moraxella (Branhamella) catarrhalis TMP-SMZ,1 cephalosporin (second- Quinolone,3 macrolide4
or third-generation)2
  Neisseria gonorrhoeae Ceftriaxone, cefixime Spectinomycin, azithromycin
  Neisseria meningitidis Penicillin G Chloramphenicol, ceftriaxone, cefotaxime
Gram-negative rods (aerobic)
  E coli, Klebsiella, Proteus Cephalosporin (first- or second- Quinolone,3 aminoglycoside5
generation),2 TMP-SMZ1
  Enterobacter, Citrobacter, Serratia TMP-SMZ,1 quinolone,3 carbapenem6 Antipseudomonal penicillin,7 aminoglycoside,5 cefepime
  Shigella Quinolone3 TMP-SMZ,1 ampicillin, azithromycin, ceftriaxone
3
  Salmonella Quinolone, ceftriaxone Chloramphenicol, ampicillin, TMP-SMZ1
  Campylobacter jejuni Erythromycin or azithromycin Tetracycline, quinolone3
  Brucella species Doxycycline + rifampin or Chloramphenicol + aminoglycoside5 or TMP-SMZ1
5
aminoglycoside
  Helicobacter pylori Proton pump inhibitor + amoxicillin Bismuth + metronidazole + tetracycline + proton pump
+ clarithromycin inhibitor
  Vibrio species Tetracycline Quinolone,3 TMP-SMZ1
  Pseudomonas aeruginosa Antipseudomonal penicillin + Antipseudomonal penicillin ± quinolone,3 cefepime,
aminoglycoside5 ceftazidime, antipseudomonal carbapenem,6 or aztreonam ±
aminoglycoside5
Burkholderia cepacia (formerly
  TMP-SMZ1 Ceftazidime, chloramphenicol
Pseudomonas cepacia)
Stenotrophomonas maltophilia (formerly
  TMP-SMZ1 Minocycline, ticarcillin-clavulanate, tigecycline, ceftazidime,
Xanthomonas maltophilia) quinolone3
  Legionella species Azithromycin or quinolone3 Clarithromycin, erythromycin
Gram-positive cocci (aerobic)
  Streptococcus pneumoniae Penicillin8 Doxycycline, ceftriaxone, antipneumococcal quinolone,3
macrolide,4 linezolid
  Streptococcus pyogenes (group A) Penicillin, clindamycin Erythromycin, cephalosporin (first-generation)2
5
  Streptococcus agalactiae (group B) Penicillin (± aminoglycoside ) Vancomycin
  Viridans streptococci Penicillin Cephalosporin (first- or third-generation),2 vancomycin
  Staphylococcus aureus    
2
  β-Lactamase negative Penicillin Cephalosporin (first-generation), vancomycin
9
  β-Lactamase positive Penicillinase-resistant penicillin As above
  Methicillin-resistant Vancomycin TMP-SMZ,1 minocycline, linezolid, daptomycin, tigecycline
10 5
  Enterococcus species Penicillin + aminoglycoside Vancomycin + aminoglycoside5
Gram-positive rods (aerobic)
  Bacillus species (non-anthracis) Vancomycin Imipenem, quinolone,3 clindamycin
1
  Listeria species Ampicillin (± aminoglycoside5) TMP-SMZ
1
  Nocardia species Sulfadiazine, TMP-SMZ Minocycline, imipenem, amikacin, linezolid
Anaerobic bacteria
 Gram-positive (clostridia, Peptococcus, Penicillin, clindamycin Vancomycin, carbapenem,6 chloramphenicol
Actinomyces, Peptostreptococcus)
  Clostridium difficile Metronidazole Vancomycin, bacitracin
  Bacteroides fragilis Metronidazole Chloramphenicol, carbapenem,6 β-lactam—β-lactamase-
inhibitor combinations, clindamycin
Fusobacterium, Prevotella,
  Metronidazole, clindamycin, As for B fragilis
Porphyromonas penicillin
(Continued)
908    SECTION VIII  Chemotherapeutic Drugs

TABLE 51–1  Empiric antimicrobial therapy based on microbiologic etiology. (Continued)


Suspected or Proven Disease or Pathogen Drugs of First Choice Alternative Drugs

Mycobacteria
  Mycobacterium tuberculosis Isoniazid + rifampin + ethambutol + Streptomycin, moxifloxacin, amikacin, ethionamide,
pyrazinamide cycloserine, PAS, linezolid
  Mycobacterium leprae    
  Multibacillary Dapsone + rifampin + clofazimine  
  Paucibacillary Dapsone + rifampin  
Mycoplasma pneumoniae Tetracycline, erythromycin Azithromycin, clarithromycin, quinolone3
Chlamydia
  C trachomatis Tetracycline, azithromycin Clindamycin, ofloxacin
  C pneumoniae Tetracycline, erythromycin Clarithromycin, azithromycin
  C psittaci Tetracycline Chloramphenicol
Spirochetes
  Borrelia recurrentis Doxycycline Erythromycin, chloramphenicol, penicillin
  Borrelia burgdorferi    
  Early Doxycycline, amoxicillin Cefuroxime axetil, penicillin
  Late Ceftriaxone  
  Leptospira species Penicillin Tetracycline
  Treponema species Penicillin Tetracycline, azithromycin, ceftriaxone
Fungi
  Aspergillus species Voriconazole Amphotericin B, itraconazole, caspofungin, isavuconazole
  Blastomyces species Amphotericin B Itraconazole, fluconazole
  Candida species Amphotericin B, echinocandin11 Fluconazole, itraconazole, voriconazole
  Cryptococcus neoformans Amphotericin B ± flucytosine (5-FC) Fluconazole, voriconazole
  Coccidioides immitis Amphotericin B Fluconazole, itraconazole, voriconazole, posaconazole
  Histoplasma capsulatum Amphotericin B Itraconazole
  Mucoraceae (Rhizopus, Absidia) Amphotericin B Posaconazole, isavuconazole
  Sporothrix schenckii Amphotericin B Itraconazole
1
Trimethoprim-sulfamethoxazole (TMP-SMZ) is a mixture of one part trimethoprim plus five parts sulfamethoxazole.
2
First-generation cephalosporins: cefazolin for parenteral administration; cefadroxil or cephalexin for oral administration. Second-generation cephalosporins: cefuroxime for par-
enteral administration; cefaclor, cefuroxime axetil, cefprozil for oral administration. Third-generation cephalosporins: ceftazidime, cefotaxime, ceftriaxone for parenteral admin-
istration; cefixime, cefpodoxime, ceftibuten, cefdinir, cefditoren for oral administration. Fourth-generation cephalosporin: cefepime for parenteral administration. Cephamycins:
cefoxitin and cefotetan for parenteral administration.
3
Quinolones: ciprofloxacin, gemifloxacin, levofloxacin, moxifloxacin, norfloxacin, ofloxacin. Norfloxacin is not effective for the treatment of systemic infections. Gemifloxacin,
levofloxacin, and moxifloxacin have excellent activity against pneumococci. Ciprofloxacin and levofloxacin have good activity against Pseudomonas aeruginosa.
4
Macrolides: azithromycin, clarithromycin, dirithromycin, erythromycin.
5
Generally, streptomycin and gentamicin are used to treat infections with Gram-positive organisms, whereas gentamicin, tobramycin, and amikacin are used to treat infections
with Gram-negatives.
6
Carbapenems: doripenem, ertapenem, imipenem, meropenem. Ertapenem lacks activity against enterococci, Acinetobacter, and P aeruginosa.
7
Antipseudomonal penicillin: piperacillin, piperacillin/tazobactam, ticarcillin/clavulanic acid.
8
See footnote 3 in Table 51–2 for guidelines on the treatment of penicillin-resistant pneumococcal meningitis.
9
Parenteral nafcillin or oxacillin; oral dicloxacillin.
10
There is no regimen that is reliably bactericidal for vancomycin-resistant enterococcus for which there is extensive clinical experience; daptomycin has bactericidal activity in
vitro. Regimens that have been reported to be efficacious include nitrofurantoin (for urinary tract infection); potential regimens for bacteremia include daptomycin, linezolid, and
dalfopristin/quinupristin.
11
Echinocandins: anidulafungin, caspofungin, micafungin.
CHAPTER 51  Clinical Use of Antimicrobial Agents     909

TABLE 51–2  Empiric antimicrobial therapy based on site of infection.


Presumed Site of Infection Common Pathogens Drugs of First Choice Alternative Drugs

Bacterial endocarditis
 Acute Staphylococcus aureus Vancomycin + ceftriaxone Penicillinase-resistant penicillin1 +
gentamicin
 Subacute Viridans streptococci, enterococci Penicillin + gentamicin Vancomycin + gentamicin
Septic arthritis
 Child Haemophilus influenzae, S aureus, Vancomycin + ceftriaxone Vancomycin + ampicillin-sulbactam or
β-hemolytic streptococci ertapenem
 Adult S aureus, Enterobacteriaceae, Vancomycin + ceftriaxone Vancomycin + ertapenem, or quinolone
Neisseria gonorrhoeae
Acute otitis media, H influenzae, Streptococcus Amoxicillin Amoxicillin-clavulanate, cefuroxime
sinusitis pneumoniae, Moraxella catarrhalis axetil, TMP-SMZ
Cellulitis S aureus, group A streptococcus Penicillinase-resistant penicillin, Vancomycin, clindamycin, linezolid,
cephalosporin (first-generation)2 daptomycin
Meningitis
 Neonate Group B streptococcus, Escherichia Ampicillin + cephalosporin Ampicillin + aminoglycoside,
coli, Listeria (third-generation) chloramphenicol, meropenem
 Child H influenzae, pneumococcus, Ceftriaxone or cefotaxime ± Chloramphenicol, meropenem
meningococcus vancomycin3
 Adult Pneumococcus, meningococcus Ceftriaxone, cefotaxime Vancomycin + ceftriaxone or cefotaxime3
Peritonitis due to Coliforms, Bacteroides fragilis Metronidazole + cephalosporin Carbapenem, tigecycline
ruptured viscus (third-generation), piperacillin/
tazobactam
Pneumonia
 Neonate As in neonatal meningitis    
 Child Pneumococcus, S aureus, Ceftriaxone, cefuroxime, Ampicillin-sulbactam
H influenzae cefotaxime
 Adult Pneumococcus, Mycoplasma, Outpatient: Macrolide,4 Outpatient: Quinolone
(community-acquired) Legionella, H influenzae, S aureus, amoxicillin, tetracycline
Chlamydophila pneumonia,
coliforms
    Inpatient: Macrolide4 + Inpatient: Doxycycline + cefotaxime,
cefotaxime, ceftriaxone, ceftriaxone, ertapenem, or ampicillin;
ertapenem, or ampicillin respiratory quinolone5
Septicemia6 Any Vancomycin + cephalosporin (third-generation) or piperacillin/tazobactam or
imipenem or meropenem
Septicemia with Any Antipseudomonal penicillin + aminoglycoside; ceftazidime; cefepime;
granulocytopenia imipenem or meropenem; consider addition of systemic antifungal therapy if
fever persists beyond 5 days of empiric therapy
1
See footnote 9, Table 51–1.
2
See footnote 2, Table 51–1.
3
When meningitis with penicillin-resistant pneumococcus is suspected, empiric therapy with this regimen is recommended.
4
Erythromycin, clarithromycin, or azithromycin (an azalide) may be used.
5
Quinolones used to treat pneumonococcal infections include levofloxacin, moxifloxacin, and gemifloxacin.
6
Adjunctive immunomodulatory drugs such as drotrecogin-alfa can also be considered for patients with severe sepsis.

determine the cause of failure. Errors in susceptibility testing are can be done to maximize it. For example, are adequate numbers
rare, but the original results should be confirmed by repeat test- of granulocytes present and is undiagnosed immunodeficiency,
ing. Drug dosing and absorption should be scrutinized and tested malignancy, or malnutrition present? The presence of abscesses
directly using serum measurements, pill counting, or directly or foreign bodies should also be considered. Finally, culture and
observed therapy. susceptibility testing should be repeated to determine whether
The clinical data should be reviewed to determine whether superinfection has occurred with another organism or whether the
the patient’s immune function is adequate and, if not, what original pathogen has developed drug resistance.
910    SECTION VIII  Chemotherapeutic Drugs

ANTIMICROBIAL (eg, β-lactams and vancomycin). For drugs whose killing action is
concentration-dependent, the rate and extent of killing increase
PHARMACODYNAMICS with increasing drug concentrations. Concentration-dependent
The time course of drug concentration is closely related to the killing is one of the pharmacodynamic factors responsible for the
antimicrobial effect at the site of infection and to any toxic effects. efficacy of once-daily dosing of aminoglycosides. For drugs whose
Pharmacodynamic factors include pathogen susceptibility testing, killing action is time-dependent, bactericidal activity continues as
drug bactericidal versus bacteriostatic activity, drug synergism, long as serum concentrations are greater than the MBC.
antagonism, and postantibiotic effects. Together with pharmaco-
kinetics, pharmacodynamic information permits the selection of Postantibiotic Effect
optimal antimicrobial dosage regimens. Persistent suppression of bacterial growth after limited exposure to
an antimicrobial agent is known as the postantibiotic effect (PAE).
Bacteriostatic versus Bactericidal Activity The PAE can be expressed mathematically as follows:
Antibacterial agents may be classified as bacteriostatic or bacte-
PAE = T – C
ricidal (Table 51–3). For agents that are primarily bacteriostatic,
inhibitory drug concentrations are much lower than bactericidal where T is the time required for the viable count in the test (in
drug concentrations. In general, cell wall-active agents are bacte- vitro) culture to increase tenfold above the count observed imme-
ricidal, and drugs that inhibit protein synthesis are bacteriostatic. diately before drug removal and C is the time required for the
The classification of antibacterial agents as bactericidal or bac- count in an untreated culture to increase tenfold above the count
teriostatic has limitations. Some agents that are considered to be observed immediately after completion of the same procedure
bacteriostatic may be bactericidal against selected organisms. On used on the test culture. The PAE reflects the time required for
the other hand, enterococci are inhibited but not killed by vanco- bacteria to return to logarithmic growth.
mycin, penicillin, or ampicillin used as single agents. Proposed mechanisms include (1) slow recovery after reversible
Bacteriostatic and bactericidal agents are equivalent for the nonlethal damage to cell structures; (2) persistence of the drug at
treatment of most infectious diseases in immunocompetent a binding site or within the periplasmic space; and (3) the need
hosts. Bactericidal agents should be selected over bacteriostatic to synthesize new enzymes before growth can resume. Most anti-
ones in circumstances in which local or systemic host defenses microbials possess significant in vitro PAEs (≥1.5 hours) against
are impaired. Bactericidal agents are required for treatment of susceptible Gram-positive cocci (Table 51–4). Antimicrobials
endocarditis and other endovascular infections, meningitis, and
infections in neutropenic cancer patients.
Bactericidal agents can be divided into two groups: agents that
exhibit concentration-dependent killing (eg, aminoglycosides TABLE 51–4  Antibacterial agents with in vitro
and quinolones) and agents that exhibit time-dependent killing postantibiotic effects ≥1.5 hours.

Against Gram-Positive Cocci Against Gram-Negative Bacilli

Aminoglycosides Aminoglycosides
TABLE 51–3  Bactericidal and bacteriostatic
Carbapenems Carbapenems
antibacterial agents.
Cephalosporins Chloramphenicol
Bactericidal Agents Bacteriostatic Agents Chloramphenicol Quinolones
Aminoglycosides Chloramphenicol Clindamycin Rifampin
Bacitracin Clindamycin Daptomycin Tetracyclines
β-Lactam antibiotics Ethambutol Glycopeptide antibiotics Tigecycline
Daptomycin Macrolides Ketolides  
Fosfomycin Nitrofurantoin Macrolides  
Glycopeptide antibiotics Novobiocin Oxazolidinones  
Isoniazid Oxazolidinones Penicillins  
Ketolides Sulfonamides Quinolones  
Metronidazole Tetracyclines Rifampin  
Polymyxins Tigecycline Streptogramins  
Pyrazinamide Trimethoprim Sulfonamides  
Quinolones   Tetracyclines  
Rifampin   Tigecycline  
Streptogramins   Trimethoprim  
CHAPTER 51  Clinical Use of Antimicrobial Agents     911

with significant PAEs against susceptible Gram-negative bacilli are may result in decreased elimination. Table 51–5 lists drugs that
limited to carbapenems and agents that inhibit protein or DNA require dosage reduction in patients with renal or hepatic insuf-
synthesis. ficiency. Failure to reduce antimicrobial agent dosage in such
In vivo PAEs are usually much longer than in vitro PAEs. patients may cause toxic effects. Conversely, patients with burns,
This is thought to be due to postantibiotic leukocyte enhance- cystic fibrosis, or trauma may have increased dosage requirements
ment (PALE) and exposure of bacteria to subinhibitory antibiotic for selected agents. The pharmacokinetics of antimicrobials is
concentrations. The efficacy of once-daily dosing regimens is in also altered in the elderly (see Chapter 60), in neonates (see
part due to the PAE. Aminoglycosides and quinolones possess Chapter 59), and in pregnancy.
concentration-dependent PAEs; thus, high doses of aminoglyco-
sides given once daily result in enhanced bactericidal activity and
extended PAEs. This combination of pharmacodynamic effects Drug Concentrations in Body Fluids
allows aminoglycoside serum concentrations that are below the Most antimicrobial agents are well distributed to most body
MICs of target organisms to remain effective for extended periods tissues and fluids. Penetration into the cerebrospinal fluid is an
of time. exception. Most do not penetrate uninflamed meninges to an
appreciable extent. In the presence of meningitis, however, the
cerebrospinal fluid concentrations of many antimicrobials increase
PHARMACOKINETIC CONSIDERATIONS (Table 51–6).

Route of Administration
Monitoring Serum Concentrations of
Many antimicrobial agents have similar pharmacokinetic proper-
ties when given orally or parenterally (ie, tetracyclines, trime-
Antimicrobial Agents
thoprim-sulfamethoxazole, quinolones, metronidazole, clindamycin, For most antimicrobial agents, the relation between dose and
rifampin, linezolid, and fluconazole). In most cases, oral therapy therapeutic outcome is well established, and serum concentra-
with these drugs is equally effective, is less costly, and results in fewer tion monitoring is unnecessary for these drugs. To justify routine
complications than parenteral therapy. serum concentration monitoring, it should be established (1)
The intravenous route is preferred in the following situations: that a direct relationship exists between drug concentrations
(1) for critically ill patients; (2) for patients with bacterial menin- and efficacy or toxicity; (2) that substantial interpatient vari-
gitis or endocarditis; (3) for patients with nausea, vomiting, gas- ability exists in serum concentrations on standard doses; (3) that
trectomy, ileus, or diseases that may impair oral absorption; and a small difference exists between therapeutic and toxic serum
(4) when giving antimicrobials that are poorly absorbed following concentrations; (4) that the clinical efficacy or toxicity of the
oral administration. drug is delayed or difficult to measure; and (5) that an accurate
assay is available.
Conditions That Alter Antimicrobial In clinical practice, serum concentration monitoring is rou-
tinely performed on patients receiving aminoglycosides or van-
Pharmacokinetics comycin. Flucytosine serum concentration monitoring has been
Various diseases and physiologic states alter the pharmacokinetics shown to reduce toxicity when doses are adjusted to maintain peak
of antimicrobial agents. Impairment of renal or hepatic function concentrations below 100 mcg/mL.

TABLE 51–5  Antimicrobial agents that require dosage adjustment or are contraindicated in patients with renal
or hepatic impairment.

Contraindicated in Dosage Adjustment Needed in


Dosage Adjustment Needed in Renal Impairment Renal Impairment Hepatic Impairment

Acyclovir, amantadine, aminoglycosides, aztreonam, Cidofovir, methenamine, Abacavir, atazanavir, caspofungin,


carbapenems, cephalosporins,1 clarithromycin, colistin, nalidixic acid, nitrofurantoin, chloramphenicol, clindamycin,
cycloserine, dalbavancin, daptomycin, didanosine, emtricitabine, sulfonamides (long-acting), erythromycin, fosamprenavir, indinavir,
ethambutol, ethionamide, famciclovir, fluconazole, tetracyclines4 metronidazole, rimantadine, tigecycline
flucytosine, foscarnet, ganciclovir, lamivudine, oseltamivir,
penicillins,2 peramivir, polymyxin B, pyrazinamide, quinolones,3
ribavirin, rifabutin, rimantadine, stavudine, telavancin,
telbivudine, telithromycin, tenofovir, terbinafine, trimethoprim-
sulfamethoxazole, valacyclovir, vancomycin, zidovudine
1
Except ceftriaxone.
2
Except antistaphylococcal penicillins (eg, nafcillin and dicloxacillin).
3
Except moxifloxacin.
4
Except doxycycline and minocycline.
912    SECTION VIII  Chemotherapeutic Drugs

TABLE 51–6  Cerebrospinal fluid (CSF) penetration of ■■ ANTIMICROBIAL DRUG


selected antimicrobials.
COMBINATIONS
CSF Concentration CSF Concentration
(Uninflamed
Meninges) as
(Inflamed
Meninges) as
RATIONALE FOR COMBINATION
Antimicrobial % of Serum % of Serum ANTIMICROBIAL THERAPY
Agent Concentration Concentration

Ampicillin 2–3 2–100


Most infections should be treated with a single antimicrobial
agent. Although indications for combination therapy exist, anti-
Aztreonam 2 5
microbial combinations are often overused in clinical practice.
Cefepime 0–2 4–12 The unnecessary use of antimicrobial combinations increases tox-
Cefotaxime 22.5 27–36 icity and costs and may occasionally result in reduced efficacy due
Ceftazidime 0.7 20–40 to antagonism of one drug by another. Antimicrobial combina-
Ceftriaxone 0.8–1.6 16 tions should be selected for one or more of the following reasons:
Cefuroxime 20 17–88 1. To provide broad-spectrum empiric therapy in seriously ill
Ciprofloxacin 6–27 26–37 patients.
Imipenem 3.1 11–41 2. To treat polymicrobial infections (such as intra-abdominal
Meropenem 0–7 1–52 abscesses, which typically are due to a combination of anaero-
bic and aerobic Gram-negative organisms, and enterococci).
Nafcillin 2–15 5–27
The antimicrobial combination chosen should cover the most
Penicillin G 1–2 8–18
common known or suspected pathogens but need not cover all
Sulfamethoxazole 40 12–47 possible pathogens. The availability of antimicrobials with
Trimethoprim <41 12–69 excellent polymicrobial coverage (eg, β-lactamase inhibitor
Vancomycin 0 1–53 combinations or carbapenems) may reduce the need for com-
bination therapy in the setting of polymicrobial infections.
3. To decrease the emergence of resistant strains. The value of
■■ MANAGEMENT OF combination therapy in this setting has been clearly demon-
strated for tuberculosis.
ANTIMICROBIAL DRUG TOXICITY 4. To decrease dose-related toxicity by using reduced doses of one
Owing to the large number of antimicrobials available, it is usually or more components of the drug regimen. The use of flucyto-
possible to select an effective alternative in patients who develop sine in combination with amphotericin B for the treatment of
serious drug toxicity (Table 51–1). However, for some infections cryptococcal meningitis in non-HIV-infected patients allows
there are no effective alternatives to the drug of choice. For example, for a reduction in amphotericin B dosage with decreased
in patients with neurosyphilis who have a history of anaphylaxis to amphotericin B–induced nephrotoxicity.
penicillin, it is necessary to perform skin testing and desensitization 5. To obtain enhanced inhibition or killing. This use of antimi-
to penicillin. It is important to obtain a clear history of drug allergy crobial combinations is discussed in the paragraphs that follow.
and other adverse drug reactions. A patient with a documented
antimicrobial allergy should carry a card with the name of the drug
and a description of the reaction. Cross-reactivity between penicil- SYNERGISM & ANTAGONISM
lins and cephalosporins is less than 10%. Cephalosporins may be
When the inhibitory or killing effects of two or more antimicrobi-
administered to patients with penicillin-induced maculopapular
als used together are significantly greater than expected from their
rashes but should be avoided in patients with a history of penicillin-
effects when used individually, synergism is said to result. Syner-
induced immediate hypersensitivity reactions. On the other hand,
gism is marked by a fourfold or greater reduction in the MIC or
aztreonam does not cross-react with penicillins and can be safely
MBC of each drug when used in combination versus when used
administered to patients with a history of penicillin-induced ana-
alone. Antagonism occurs when the combined inhibitory or kill-
phylaxis. For mild reactions, it may be possible to continue therapy
ing effects of two or more antimicrobial drugs are significantly less
with use of adjunctive agents or dosage reduction.
than observed when the drugs are used individually.
Adverse reactions to antimicrobials occur with increased fre-
quency in several groups, including neonates, geriatric patients,
renal failure patients, and AIDS patients. Dosage adjustment of Mechanisms of Synergistic Action
the drugs listed in Table 51–5 is essential for the prevention of The need for synergistic combinations of antimicrobials has been
adverse effects in patients with renal failure. In addition, several clearly established for the treatment of enterococcal endocarditis.
agents are contraindicated in patients with renal impairment Bactericidal activity is essential for the optimal management of
because of increased rates of serious toxicity (Table 51–5). See the bacterial endocarditis. Penicillin or ampicillin in combination
preceding chapters for discussions of specific drugs. with gentamicin or streptomycin is superior to monotherapy with
CHAPTER 51  Clinical Use of Antimicrobial Agents     913

a penicillin or vancomycin. When tested alone, penicillins and antagonize the action of bactericidal cell wall-active agents
vancomycin are only bacteriostatic against susceptible enterococ- because cell wall-active agents require that the bacteria be
cal isolates. When these agents are combined with an amino- actively growing and dividing.
glycoside, however, bactericidal activity results. The addition of 2. Induction of enzymatic inactivation: Some Gram-negative bacilli,
gentamicin or streptomycin to penicillin allows for a reduction in including Enterobacter species, P aeruginosa, Serratia marcescens,
the duration of therapy for selected patients with viridans strepto- and Citrobacter freundii, possess inducible β-lactamases. β-Lactam
coccal endocarditis. antibiotics such as imipenem, cefoxitin, and ampicillin are potent
Other synergistic antimicrobial combinations have been shown inducers of β-lactamase production. If an inducing agent is com-
to be more effective than monotherapy with individual compo- bined with an intrinsically active but hydrolyzable β-lactam such
nents. Trimethoprim-sulfamethoxazole has been successfully used as piperacillin, antagonism may result.
in the treatment of bacterial infections and P jiroveci (carinii)
pneumonia.* β-Lactamase inhibitors restore the activity of intrin-
sically active but hydrolyzable β lactams against organisms such as
Staphylococcus aureus and Bacteroides fragilis. Three major mecha- ■■ ANTIMICROBIAL
nisms of antimicrobial synergism have been established: PROPHYLAXIS
1. Blockade of sequential steps in a metabolic sequence:
Antimicrobial agents are effective in preventing infections in
Trimethoprim-sulfamethoxazole is the best-known example of
many settings. Antimicrobial prophylaxis should be used in cir-
this mechanism of synergy (see Chapter 46). Blockade of the
cumstances in which efficacy has been demonstrated and benefits
two sequential steps in the folic acid pathway by trimethoprim-
outweigh the risks of prophylaxis. Antimicrobial prophylaxis may
sulfamethoxazole results in a much more complete inhibition
be divided into surgical prophylaxis and nonsurgical prophylaxis.
of growth than achieved by either component alone.
2. Inhibition of enzymatic inactivation: Enzymatic inactivation
of β-lactam antibiotics is a major mechanism of antibiotic Surgical Prophylaxis
resistance. Inhibition of β lactamase by β-lactamase inhibitor Surgical wound infections are a major category of nosocomial
drugs (eg, sulbactam) results in synergism. infections. The estimated annual cost of surgical wound infections
3. Enhancement of antimicrobial agent uptake: Penicillins and in the USA is more than $1.5 billion.
other cell wall-active agents can increase the uptake of amino- The National Research Council (NRC) Wound Classification
glycosides by a number of bacteria, including staphylococci, Criteria have served as the basis for recommending antimicrobial
enterococci, streptococci, and P aeruginosa. Enterococci are prophylaxis. NRC criteria consist of four classes (see Box: National
thought to be intrinsically resistant to aminoglycosides because Research Council [NRC] Wound Classification Criteria).
of permeability barriers. Similarly, amphotericin B is thought The Study of the Efficacy of Nosocomial Infection Control
to enhance the uptake of flucytosine by fungi. (SENIC) identified four independent risk factors for postop-
erative wound infections: operations on the abdomen, operations
lasting more than 2 hours, contaminated or dirty wound classifi-
Mechanisms of Antagonistic Action cation, and at least three medical diagnoses. Patients with at least
There are few clinically relevant examples of antimicrobial two SENIC risk factors who undergo clean surgical procedures
antagonism. The most striking example was reported in a study of have an increased risk of developing surgical wound infections and
patients with pneumococcal meningitis. Patients who were treated should receive antimicrobial prophylaxis.
with the combination of penicillin and chlortetracycline had a Surgical procedures that necessitate the use of antimicrobial
mortality rate of 79% compared with a mortality rate of 21% in prophylaxis include contaminated and clean-contaminated opera-
patients who received penicillin monotherapy (illustrating the first tions, selected operations in which postoperative infection may
mechanism set forth below). be catastrophic such as open heart surgery, clean procedures that
The use of an antagonistic antimicrobial combination does involve placement of prosthetic materials, and any procedure
not preclude other potential beneficial interactions. For example, in an immunocompromised host. The operation should carry a
rifampin may antagonize the action of anti-staphylococcal penicil- significant risk of postoperative site infection or cause significant
lins or vancomycin against staphylococci. However, the aforemen- bacterial contamination.
tioned antimicrobials may prevent the emergence of resistance to General principles of antimicrobial surgical prophylaxis include
rifampin. the following:
Two major mechanisms of antimicrobial antagonism have been 1. The antibiotic should be active against common surgical
established: wound pathogens; unnecessarily broad coverage should be
1. Inhibition of cidal activity by static agents: Bacteriostatic avoided.
agents such as tetracyclines and chloramphenicol can 2. The antibiotic should have proved efficacy in clinical trials.
3. The antibiotic must achieve concentrations greater than the
*
Pneumocystis jiroveci is a fungal organism found in humans (P carinii MIC of suspected pathogens, and these concentrations must be
infects animals) that responds to antiprotozoal drugs. See Chapter 52. present at the time of incision.
914    SECTION VIII  Chemotherapeutic Drugs

The proper selection and administration of antimicrobial


National Research Council (NRC) Wound prophylaxis are of utmost importance. Common indications for
Classification Criteria surgical prophylaxis are shown in Table 51–7. Cefazolin is the
prophylactic agent of choice for head and neck, gastroduodenal,
Clean: Elective, primarily closed procedure; respiratory, gas- biliary tract, gynecologic, and clean procedures. Local wound
trointestinal, biliary, genitourinary, or oropharyngeal tract infection patterns should be considered when selecting antimicro-
not entered; no acute inflammation and no break in tech- bial prophylaxis. The selection of vancomycin over cefazolin may
nique; expected infection rate ≤ 2%. be necessary in hospitals with high rates of methicillin-resistant
Clean contaminated: Urgent or emergency case that is S aureus or S epidermidis infections. The antibiotic should be pres-
otherwise clean; elective, controlled opening of respiratory, ent in adequate concentrations at the operative site before incision
gastrointestinal, biliary, or oropharyngeal tract; minimal and throughout the procedure; initial dosing is dependent on the
spillage or minor break in technique; expected infection volume of distribution, peak levels, clearance, protein binding,
rate ≤ 10%. and bioavailability. Parenteral agents should be administered dur-
Contaminated: Acute nonpurulent inflammation; major ing the interval beginning 60 minutes before incision. In cesarean
technique break or major spill from hollow organ; penetrat- section, the antibiotic is administered after umbilical cord clamp-
ing trauma less than 4 hours old; chronic open wounds to be ing. For many antimicrobial agents, doses should be repeated if
grafted or covered; expected infection rate about 20%. the procedure exceeds 2–6 hours in duration. Single-dose pro-
Dirty: Purulence or abscess; preoperative perforation of phylaxis is effective for most procedures and results in decreased
respiratory, gastrointestinal, biliary, or oropharyngeal tract; toxicity and antimicrobial resistance.
penetrating trauma more than 4 hours old; expected infec- Improper administration of antimicrobial prophylaxis leads
tion rate about 40%. to excessive surgical wound infection rates. Common errors in
antibiotic prophylaxis include selection of the wrong antibiotic,
administering the first dose too early or too late, failure to repeat
doses during prolonged procedures, excessive duration of prophy-
4. The shortest possible course—ideally a single dose—of the
laxis, and inappropriate use of broad-spectrum antibiotics.
most effective and least toxic antibiotic should be used.
5. The newer broad-spectrum antibiotics should be reserved for
therapy of resistant infections.
Nonsurgical Prophylaxis
6. If all other factors are equal, the least expensive agent should Nonsurgical prophylaxis includes the administration of antimi-
be used. crobials to prevent colonization or asymptomatic infection as

TABLE 51–7  Recommendations for surgical antimicrobial prophylaxis.


Type of Operation Common Pathogens Drug of Choice

Cardiac (with median sternotomy) Staphylococci, enteric Gram-negative rods Cefazolin


Noncardiac, thoracic Staphylococci, streptococci, enteric Gram-negative rods Cefazolin
Vascular (abdominal and lower Staphylococci, enteric Gram-negative rods Cefazolin
extremity)
Neurosurgical (craniotomy) Staphylococci Cefazolin
Orthopedic (with hardware insertion) Staphylococci Cefazolin
Head and neck (with entry into the Staphylococcus aureus, oral flora Cefazolin + metronidazole
oropharynx)
Gastroduodenal S aureus, oral flora, enteric Gram-negative rods Cefazolin
Biliary tract S aureus, enterococci, enteric Gram-negative rods Cefazolin
Colorectal (elective surgery) Enteric Gram-negative rods, anaerobes Oral erythromycin + neomycin1
Colorectal (emergency surgery or Enteric Gram-negative rods, anaerobes Cefoxitin, cefotetan, ertapenem, or
obstruction) cefazolin + metronidazole
Appendectomy, nonperforated Enteric Gram-negative rods, anaerobes Cefoxitin, cefotetan, or cefazolin +
metronidazole
Hysterectomy Enteric Gram-negative rods, anaerobes, enterococci, group B Cefazolin, cefotetan, or cefoxitin
streptococci
Cesarean section Enteric Gram-negative rods, anaerobes, enterococci, group B Cefazolin
streptococci
1
In conjunction with mechanical bowel preparation.
CHAPTER 51  Clinical Use of Antimicrobial Agents     915

TABLE 51–8  Recommendations for nonsurgical antimicrobial prophylaxis.


Infection to Be
Prevented Indication(s) Drug of Choice Efficacy

Anthrax Suspected exposure Ciprofloxacin or doxycycline Proposed effective


Cholera Close contacts of a case Tetracycline Proposed effective
Diphtheria Unimmunized contacts Penicillin or erythromycin Proposed effective
Endocarditis Dental, oral, or upper respiratory tract procedures1 in Amoxicillin or clindamycin Proposed effective
at-risk patients2
Genital herpes simplex Recurrent infection (≥4 episodes per year) Acyclovir Excellent
Perinatal herpes simplex Mothers with primary HSV or frequent recurrent Acyclovir Proposed effective
type 2 infection genital HSV
Group B streptococcal Mothers with cervical or vaginal GBS colonization and Ampicillin or penicillin Excellent
(GBS) infection their newborns with one or more of the following: (a)
onset of labor or membrane rupture before 37 weeks’ ges-
tation, (b) prolonged rupture of membranes (>12 hours),
(c) maternal intrapartum fever, (d) history of GBS bacteri-
uria during pregnancy, (e) mothers who have given birth
to infants who had early GBS disease or with a history of
streptococcal bacteriuria during pregnancy
Haemophilus influenzae Close contacts of a case in incompletely immunized Rifampin Excellent
type B infection children (>48 months old)
HIV infection Health care workers exposed to blood after needle-stick Tenofovir/emtricitabine and Good
injury raltegravir
  Pregnant HIV-infected women who are at ≥14 weeks of HAART3 Excellent
gestation; newborns of HIV-infected women for the first
6 weeks of life, beginning 8–12 hours after birth
Influenza A and B Unvaccinated geriatric patients, immunocompromised Oseltamivir Good
hosts, and health care workers during outbreaks
Malaria Travelers to areas endemic for chloroquine-susceptible Chloroquine Excellent
disease
  Travelers to areas endemic for chloroquine-resistant Mefloquine, doxycycline, or Excellent
disease atovaquone/proguanil
Meningococcal infection Close contacts of a case Rifampin, ciprofloxacin, or Excellent
ceftriaxone
Mycobacterium avium HIV-infected patients with CD4 count <75/μL Azithromycin, clarithromycin, Excellent
complex or rifabutin
Otitis media Recurrent infection Amoxicillin Good
Pertussis Close contacts of a case Azithromycin Excellent
Plague Close contacts of a case Tetracycline Proposed effective
Pneumococcemia Children with sickle cell disease or asplenia Penicillin Excellent
Pneumocystis jiroveci High-risk patients (eg, AIDS, leukemia, transplant) Trimethoprim-sulfamethoxa- Excellent
pneumonia (PCP) zole, dapsone, or atovaquone
Rheumatic fever History of rheumatic fever or known rheumatic heart Benzathine penicillin Excellent
disease
Toxoplasmosis HIV-infected patients with IgG antibody to Toxoplasma Trimethoprim- Good
and CD4 count <100/μL sulfamethoxazole
Tuberculosis Persons with positive tuberculin skin tests and one or Isoniazid or rifampin or iso- Excellent
more of the following: (a) HIV infection, (b) close contacts niazid + rifapentine
with newly diagnosed disease, (c) recent skin test conver-
sion, (d) medical conditions that increase the risk of devel-
oping tuberculosis, (e) age < 35 y
Urinary tract infections Recurrent infection Trimethoprim-sulfamethox- Excellent
(UTI) azole
1
Prophylaxis is recommended for the following: dental procedures that involve manipulation of gingival tissue or the periapical region of teeth or perforation of the oral mucosa,
and invasive procedure of the respiratory tract that involves incision or biopsy of the respiratory mucosa, such as tonsillectomy and adenoidectomy.
2
Prophylaxis should be targeted to those with the following risk factors: prosthetic heart valves, previous bacterial endocarditis, congenital cardiac malformations, cardiac trans-
plantation patients who develop cardiac valvulopathy.
3
Highly active antiretroviral therapy. See aidsinfo.nih.gov/ for updated guidelines.
916    SECTION VIII  Chemotherapeutic Drugs

well as the administration of drugs following colonization by or because of underlying disease (eg, immunocompromised hosts).
inoculation of pathogens but before the development of disease. Prophylaxis is most effective when directed against organisms
Nonsurgical prophylaxis is indicated in individuals who are at that are predictably susceptible to antimicrobial agents. Common
high risk for temporary exposure to selected virulent pathogens indications and drugs for nonsurgical prophylaxis are listed in
and in patients who are at increased risk for developing infection Table 51–8.

REFERENCES Martin RF: Surgical infections. Surg Clin North Am 2014;94:1135.


Mirakian R et al: Management of allergy to penicillins and other beta-lactams.
Baddour LM et al: Infective endocarditis: Diagnosis, antimicrobial therapy, and
Clin Exp Allergy 2015;45:300.
management of complications. Circulation 2015;132:1435.
National Nosocomial Infections Surveillance System: National Nosocomial Infec-
Baron EJ et al: Guide to utilization of the microbiology laboratory for diagnosis of
tions Surveillance (NNIS) System Report, Data Summary from January
infectious diseases: 2013 recommendations by the Infectious Diseases Soci-
1992–June 2004, issued October 2004. Am J Infect Control 2004;32:470.
ety of America (IDSA) and the American Society for Microbiology (ASM).
Clin Infect Dis 2013;57:e22. Panel on Opportunistic Infections in HIV-Infected Adults and Adolescents:
Guidelines for the prevention and treatment of opportunistic infections in
Blumberg HM et al: American Thoracic Society/Centers for Disease Control and
HIV-infected adults and adolescents: Recommendations from the Centers
Prevention/Infectious Diseases Society of America: Treatment of tuberculo-
for Disease Control and Prevention, the National Institutes of Health, and
sis. Am J Respir Crit Care Med 2003;167:603.
the HIV Medicine Association of the Infectious Diseases Society of America.
Boucher HW: 10 × ‘20 Progress—Development of new drugs active against gram- Available at: https://aidsinfo.nih.gov/contentfiles/lvguidelines/adult_oi.pdf.
negative bacilli: An update from the Infectious Diseases Society of America.
Peleg AY et al: Hospital-acquired infections due to gram negative bacteria. N Engl
Clin Infect Dis 2013;56:1685.
J Med 2010;362:1804.
Bratzler DW et al: Clinical practice guidelines for antimicrobial prophylaxis in
Simons FE: Anaphylaxis. J Allergy Clin Immunol 2010;125(Suppl 2):S161.
surgery. Am J Health Syst Pharm 2013;70:195.
Spellberg B et al: The future of antibiotics and resistance. N Engl J Med
Chow AW et al: IDSA clinical practice guideline for acute bacterial rhinosinusitis
2013;368:299.
in children and adults. Clin Infect Dis 2012;54:e72.
Tunkel AR et al: Practice guidelines for the management of bacterial meningitis.
Holmes AH et al: Understanding the mechanisms and drivers of antimicrobial
Clin Infect Dis 2004;39:1267.
resistance. Lancet 2016;387:176.
Wilson W et al: Prevention of infective endocarditis: Guidelines from the
Kalil AC et al: Management of adults with hospital-acquired and ventilator-
American Heart Association. Circulation 2007;116:1736.
associated pneumonia: 2016 Clinical practice guidelines by the Infectious
Disease Society of America and the American Thoracic Society. Clin Infect Workowski KA, Bolan GA: Sexually transmitted diseases treatment guidelines
Dis 2016;63:e61. 2015. Centers for Disease Control and Prevention. MMWR Morb Mortal
Wkly Rep 2015;64(RR-3):1.
Kaye KS, Kaye D: Antibacterial therapy and newer agents. Infect Dis Clin North
Am 2009;23:757.
Mandell LA et al: Infectious Diseases Society of America/American Thoracic
Society Consensus guidelines on the management of community-acquired
pneumonia in adults. Clin Infect Dis 2007;44:S27.

C ASE STUDY ANSWER

This patient is experiencing a post-procedure urinary negative organisms such as Pseudomonas aeruginosa. The
tract infection which may have been introduced into patient was treated with vancomycin and meropenem,
his bloodstream at the time of his cystoscopy. It is likely and blood and urine cultures were both positive for
that the patient is experiencing a sepsis-like syndrome ESBL-positive E coli that was resistant to ciprofloxacin.
and has a systemic infection with a uropathogen that The patient defervesced and hemodynamically stabilized
is resistant to the antibiotic that he has received. Pos- over the subsequent 48 hours. ESBL-positive E coli is an
sible bacteria that may be responsible for the patient’s emerging urinary tract pathogen that may be acquired in
symptoms are methicillin-resistant Staphylococcus aureus, the outpatient setting, and oral antibiotic therapy may not
Enterococcus sp., and enteric Gram-negative rods that are reliably be effective; empiric therapy with a carbapenem
resistant to ciprofloxacin such as ESBL-positive E coli or (ertapenem, doripenem, meropenem, imipenem) is recom-
Klebsiella pneumoniae, or other hospital-acquired Gram mended for serious infections due to this organism.

You might also like