You are on page 1of 31

33

Pharmacology of Specific Drug


Groups: Antibiotic Therapy*
Purnima Kumar

K E Y I N F O R M AT I O N
• The major mechanisms by which antibacterial drugs act are the • Horizontal gene transfer (one microorganism acquiring DNA
following: from another microorganism) can take place by the following:
• Inhibition of cell wall synthesis • Transduction
• Alteration of cell membrane function • Transformation
• Inhibition of ribosomal protein synthesis • Conjugation
• Inhibition of DNA or RNA synthesis or function • Adverse antibiotic effects vary from class to class. Some common
• Inhibition of folic acid synthesis ones for certain classes include these:
• Mechanisms by which bacteria become resistant to antimicrobial • Allergies
drugs include these: • Superinfections, such as pseudomembranous colitis
• Enzymatic inactivation of the drug • Gastrointestinal adverse effects
• Change in the target of the drug • Renal toxicity
• Reducing permeability of the bacterial cell • Ototoxicity
• Active drug efflux from the cell
• Changing growth requirements, bypassing the mechanism of
the drug
• Overproduction of the drug target(s)

CASE STUDY to disinfect wounds. In the 1870s, Robert Koch proved the bacterial
causation of anthrax and tuberculosis, and in the 1880s, Pasteur
A 20-year-old female college sophomore presents with a complaint of developed anthrax and rabies vaccines. In 1891, Paul Ehrlich
“extremely painful gums” (see Fig. 33-1). The patient also complains of chills showed that antibodies were responsible for immunity. In 1897,
for the past two days. You record her temperature at 102 °F. Clinical exam- Ivanowski and Beiternick discovered viruses. The mosquito vector
ination reveals punched-out interdental papillae, gingival bleeding, abun- for yellow fever was described in 1900, Treponema pallidum was
dant plaque, and a fetid odor. What is the most likely diagnosis? How would found to be the cause of syphilis in 1905, human immunodeficiency
you treat this patient? Would you use systemic medications for treatment? virus (HIV) was identified in 1983, Helicobacter pylori was discov-
Describe potential and typical adverse effects of your prescription(s). ered as a cause of peptic ulcer in 1984, and the West Nile virus was
identified in 1999.
In the early 1900s, Paul Ehrlich used the term magic bullet for
his predicted chemical that would affect only microbial cells and
INTRODUCTION have no effect on mammalian cells. He later used fuchsin and mer-
The modern era of infectious disease began with the first visualization cury (Salvarsan) to treat syphilis. In 1928, Alexander Fleming ser-
of microbes by Anton van Leeuwenhoek in 1683, the “animicules” endipitously discovered that a mold, Penicillium chrysogenum, lysed
of dental plaque scraped from his upper gingiva and killed with salt staphylococci; this was later developed to its full potential by the
(the first periodontal chemotherapy). In 1776, Edward Jenner admin- isolation of penicillin from Penicillium notatum by Florey and col-
istered the first smallpox vaccination. In 1848, Ignaz Semmelweiss leagues at Oxford in the late 1930s and early 1940s. The first use of
introduced clean surgical operating technique (“gentlemen, wash your penicillin was in 1941 on an English police constable with strepto-
hands”). In 1854, John Snow showed the link between cholera and coccal and staphylococcal skin abscesses. In the United States, pen-
drinking water. icillin was first used in 1942 on Anne Miller, who had streptococcal
In the 1860s, Louis Pasteur first used the word germ for living toxemia of pregnancy. Another ground-breaking event in medical
entities that produced disease, and Joseph Lister used carbolic acid advances was the demonstration in 1935 by Gerhard Domagk that
sulfanilamide could be safely used systemically to treat infectious
disease. Thus was born the era of antibiotics and anti-microbials,
*The author wishes to recognize Dr. Thomas J. Pallasch for his past contribu- arguably one of the most revolutionary events in the history of
tions to this chapter. mankind.

457
458 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

become cidal at high concentrations. The previous preference for cidal


MECHANISMS OF ACTION
drugs over static antibiotics (cidal drugs allegedly do not rely on host
By strict definition, antibiotics are chemicals that are derived from defenses) has become less distinct because of the appreciation of the
microorganisms (commonly yeasts and fungi) and are used to inhibit long post-antibiotic effects (continued antibiotic activity when the
other microorganisms. Almost all clinically antibacterial drugs are drug blood levels have declined) of bacteriostatic drugs.
derived from naturally occurring entities, with only few that are
entirely synthetically produced (sulfonamides, fluoroquinolones, and Inhibition of Cell Wall Synthesis
oxazolidinones). These drugs are not antibiotics by the narrow defini- The principal cell wall inhibitors are β-lactam antibiotics and gly-
tion but are included in the discussion of antibiotics. copeptides. Bacterial cell walls are rigid and composed of alternat-
Antimicrobials affect the viability of microorganisms by five known ing peptidoglycan (murein) units of N-acetyl-d-glucosamine and
processes: (1) inhibition of cell wall synthesis, (2) alteration of cell N-acetylmuramic acid (NAM). These are cross-linked via short pep-
membrane integrity, (3) inhibition of ribosomal protein synthe- tides by amide linkages to a d-alanyl group on NAM. Various bacte-
sis, (4) suppression of deoxyribonucleic acid (DNA) synthesis, and rial enzymes (transglycosylases, transpeptidases, carboxypeptidases,
(5) inhibition of folic acid synthesis (Fig. 33-2). Microbial cell wall endopeptidases) catalyze the formation of the rigid cell wall by incor-
synthesis inhibition and membrane effects are extra-cytoplasmic, and porating new peptidoglycan into existing peptidoglycan and then
inhibition of nucleic acid, protein, and folic acid synthesis are intra-cy- cross-linking to form a rigid cell wall. The internal osmotic pressure
toplasmic. Drugs that affect bacterial cell wall or membrane integrity of the bacterium causes lysis of the bacterial cell because the wall is no
and DNA synthesis are usually, but not always, bactericidal (inducing longer an effective barrier (see Fig. 33-2).
cell death), and protein and folic acid synthesis inhibitors are usually In addition, in some organisms, an antibiotic may inhibit the
bacteriostatic (preventing cell growth or replication). inhibitor of an endogenous bacterial autolysin (N-acetyl-muramyl-
Whether an antimicrobial agent is bactericidal (cidal) or bacte- l-alanine amidase). The autolysin causes the lysis of the bacterial cell
riostatic (static) can also depend on its concentration at the infected wall.
site and the particular offending organism because some static drugs
Alteration in Cell Membrane Integrity
A drug may disrupt the integrity of the cell membrane by displacing
Ca2+ and Mg++ from membrane lipid phosphate groups. Cationic
antimicrobial peptides are part of humans’ natural skin and mucosal
defense system and act by disrupting cell wall or membrane integ-
rity by an effect on the gram-negative lipopolysaccharide compo-
nent that literally puts holes in the wall or membrane (see Fig. 33-2).

Inhibition of Ribosomal Protein Synthesis


Antibiotics may inhibit at either the 30S or 50S ribosomal subunit.
This can result in inhibition of initiation of protein synthesis, inhi-
bition of peptidyl transferase, and inhibition of the extrusion of the
peptide chain from the ribosome, causing misreading of the mRNA
message and other mechanisms (see Fig. 33-2).

Inhibition of Nucleic Acid Synthesis


Mechanisms associated with either RNA or DNA include inhibition
of DNA gyrase and topoisomerase IV, inhibition of DNA-dependent
FIG 33-1  Photograph of patient in the case study. RNA polymerase, and damage to DNA (see Fig. 33-2).

Inhibitors of Inhibitors of
metabolism protein synthesis
{Sulfonamides} {Tetracyclines}
{Aminoglycosides}
{Mactrolides}

THF DNA
A Ribosomes
mRNA

PAB
A

Inhibitors of nucleic
Inhibitors of cell
acid function or
wall synthesis
synthesis
{β-Lactams}
{Fluoroquinolones}
Inhibitors of cell
membrane function
{Amphotericin B}
{Isoniazid}
FIG 33-2  Mechanisms of action of antimicrobial drugs and representative drugs.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 459

Inhibition of Folic Acid Synthesis Enzymatic inactivation is one of the more common methods and
Sulfonamides and trimethoprim are anti-metabolites that inhibit is typified by β-lactamase hydrolysis of penicillins and cephalosporins
sequential steps in the bacterial synthesis of folic acid essential for and acetyltransferases that inactivate chloramphenicol, aminoglyco-
one-carbon transfers in nucleic acid synthesis (see Fig. 33-2). sides, and tetracyclines. Altered target sites include ribosomal point
mutations for tetracyclines, macrolides, and clindamycin; altered
MICROBIAL RESISTANCE TO ANTIBIOTICS DNA gyrase and topoisomerase for fluoroquinolones; and modified
penicillin-binding proteins (PBPs) for viridans group streptococci
(FIG. 33-3)
(VGS) and pneumococci. Most microorganisms have developed
Microbial resistance to antibiotics has become a major factor in deter- ways to alter their cell wall or membrane permeability to limit access
mining when and which antibiotic is used and dosages and length of of the antibiotic to its receptor by deleting outer membrane proteins
administration. It also has spurred renewed interest in antibiotic phar- or closing membrane pore channels. Altering antibiotic access to
macokinetics and pharmacodynamics. the cell interior usually does not confer a high level of resistance on
Procedures designed to reduce antibiotic-resistant pathogenic the organism and must be combined with another mechanism for
microorganisms have been developed, including education of health significant resistance potential. Several hundred efflux proteins are
care providers and the general public, improved handwashing tech- available that extrude waste products from the microbial cell but that
niques, better hospital infection control, isolation of patients with now have been adapted over time to eliminate antibiotics specifically
highly resistant bacteria, control of antibiotic use in hospitals through from the cell interior virtually as fast as they can enter. Enterococci can
formularies and pharmacist oversight, and the removal of antibiotics evade destruction by developing alternative metabolic growth require-
for growth promotion in agricultural animals. ments (auxotrophs). Sulfonamide resistance may occur from the
All microbial resistance is local; the patterns and extent of this resis- overproduction of para-aminobenzoic acid (PABA), and some enteric
tance are determined by the use of antibiotics in a particular commu- organisms evade β-lactam antibiotics by overproducing β-lactamase
nity. What is true in Miami may not be true in Los Angeles or in Paris, (hyper–β-lactamase producers).
London, Rome, or New Delhi. If tetracyclines are used widely in the Antibiotic tolerance occurs when the antibiotic no longer kills
community for acne or Lyme disease, a high resistance level to the drug the microorganism, but it merely inhibits its growth or multiplica-
is likely to be present in that locale. If not, the microbial resistance level tion. Tolerant microorganisms start to grow after the antibiotic is
is likely to be low. If an antibiotic or its analogue has been used widely in removed, whereas resistant microorganisms multiply in the presence
agriculture, this may strongly influence resistance patterns, to the point of the antibiotic. Tolerance is usually caused by the loss of autolysin
of rendering a new antibiotic far less useful. In Taiwan, virginiamycin activity through a failure to create or mobilize the autolytic enzymes.
(a streptogramin) has been used for more than 2 decades as a growth Vancomycin tolerance in Streptococcus pneumoniae is unique; a muta-
promoter in food animals. When quinupristin-dalfopristin, a new strep- tion in the sensor-response system controls the bactericidal autolysin
togramin, was tested on human bacterial isolates before its clinical intro- activity.
duction, more than 50% of some pathogens were already resistant to A major factor in the development and maintenance of antibiotic
the drug. Antibiotics are truly societal drugs that cumulatively affect the resistance in microbes is that sensitive microorganisms are inhibited,
individual receiving the drug and many others as well. allowing resistant ones to multiply and dominate. Microbial resistance
Microorganisms have developed six known mechanisms to evade is most likely to occur when subtherapeutic antibiotic doses are used;
the bactericidal or bacteriostatic actions of antimicrobials, as follows: these are doses that do not kill or inhibit the microorganism, but rather
(1) enzymatic inactivation, (2) modification/protection of the target allow it to perceive the chemical as a threat to its survival and to react
site, (3) limited access of antibiotic (altered cell membrane permea- by mutation to resistance, acquisition, or transfer of resistance genes/
bility), (4) active drug efflux, (5) use of alternative growth require- virulence factors or induction (expression) of latent resistance genes.
ments, and (6) overproduction of target sites (see Fig. 33-3). The gastrointestinal tract is a massive reservoir for resistance genes
readily transferred within and between enteric microbial species, a
process greatly enhanced by antibiotics that readily induce the expres-
sion or transfer of resistance genes.

Alteration of target sites


SPECIFIC RESISTANCE MECHANISMS
Alternate growth requirements β-Lactamases
The most important acquired mechanism for β-lactam resistance,
particularly in gram-negative microorganisms, is the production of
various β-lactamases that hydrolyze the β-lactam ring to form a linear
Enzymatic inactivation
metabolite incapable of binding to PBPs.
Overproduction of target sites
β-Lactamases have been variously classified by Richmond-Sykes
(I to V), Ambler (A to D), and Bush (1 to 4). β-Lactamase enzymes
can be chromosomally mediated or easily transferred by transpos-
able elements. The most pressing difficulties with β-lactamases are
their widespread dissemination throughout the microbial environ-
Efflux pumps ment, ability to move between widely disparate organisms, tendency
to inhibit new antibiotic agents rapidly, and increasing resistance
to β-lactamase inhibitors (clavulanic acid, sulbactam, tazobactam,
Decrease in cell permeability and avibactam). β-Lactamases have been observed in numerous
FIG 33-3  Mechanisms by which bacteria can become resistant to anti- gram-positive and gram-negative pathogens. The cohabitation of
microbial drugs. staphylococci and enterococci on human skin in hospitals has likely
460 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

led to the incorporation of β-lactamases genes into enterococci after


TABLE 33-1  Antibiotic Resistance Patterns
the latter organisms had successfully avoided this transfer for billions
of years. Antimicrobial Drugs and Examples of
Point mutations have appeared more recently in certain β-lac- Mechanisms Mechanisms
tamases, resulting in extended-spectrum β-lactamases in Klebsiella Enzymatic ­antibiotic β-Lactams by β-lactamases
pneumoniae that hydrolyze the latest cephalosporins. Certain enteric inactivation Aminoglycosides by aminoglycoside-modifying enzymes
microorganisms (Escherichia coli, Citrobacter freundii, K. pneumoniae, Chloramphenicol by acetyltransferases
Proteus mirabilis) can produce massive amounts of TEM-1 ­β-lactamase Streptogramins by acetyltransferases
(hyper–β-lactamase producers) that can overwhelm β-lactamase Tetracyclines by enzymatic oxidation
inhibitors. Metallo-β-lactamases possess the broadest spectrum of Modification/­ β-Lactams: altered PBPs
inhibitory activity and hydrolyze all β-lactam antibiotics except mono- protection of Fluoroquinolones: altered DNA gyrase or topoisomerases
bactams (aztreonam) and are not inhibited by any of the β-lactamase target site Rifampin: altered RNA polymerase
inhibitors currently available. Sulfonamides: altered dihydropteroate synthase
The first plasmid-encoded β-lactamases with the ability to hydro- Trimethoprim: altered dihydrofolate reductase
lyze cephalosporins were termed the extended-spectrum β-lactamases Macrolide-lincosamide-streptogramin B aggregate
(ESBLs). These ESBL microbes were also resistant to aminoglycosides, gene (MLSb): methylation of adenine on 23S rRNA
tetracyclines, and trimethoprim/sulfonamides. ESBLs cause resistance Glycopeptides: change of d-Ala-d-Ala to d-Ala-­d-
to third-generation cephalosporins (cefotaxime, ceftriaxone, ceftazi- lactate in cell wall
dime) and monobactams (aztreonam) but are sensitive to cephamy- Tetracyclines: ribosomal protection
cins (cefoxitin, cefotetan) and carbapenems (imipenem, meropenem, Limiting access of β-Lactams, fluoroquinolones, most antibiotics: altered
ertapenem). These ESBL microbes are horizontally transmitted by antibiotic outer membrane porins
mobile genetic elements from food, animals, or family members and Most antibiotics: reduced membrane transport
induce greater mortality than enteric bacilli without these ESBLs. Active antibiotic Tetracyclines: TET genes
efflux Fluoroquinolones: Nor A genes
Multidrug Antibiotic Efflux Pumps Failure to activate Metronidazole: decreased flavodoxin production
Certain bacteria can move an antibiotic out of the cell as soon as it antibiotic
enters. Currently, more than 50 such systems have been described, Use of alternative Enterococcal auxotrophs
and these cytoplasmic membrane transport proteins (multidrug efflux growth requirements
pumps) have likely evolved to protect the cell from foreign chemi- Overproduction of Sulfonamides: overproduction of PABA
cal invasion and allow its secretion of cell metabolic products. Efflux target sites Enteric bacilli: overproduction of β-lactamase
pumps operate in E. coli, Pseudomonas aeruginosa, staphylococci,
Streptococcus pyogenes, S. pneumoniae, Bacillus subtilis, Pasteurella mul­ PABA, p-Aminobenzoic acid; PBPs, penicillin-binding proteins.
tocida, Neisseria gonorrhoeae, mycobacteria, and enterococci. For tet- From Polk R: Optimal use of modern antibiotics: emerging trends, Clin
Infect Dis 29:264-274, 1999; Smith H: Host factors that influence the
racyclines, these efflux pumps are the major mechanism for resistance
behavior of bacteria pathogens in vivo, Int J Med Microbiol 290:207-
and are becoming increasingly so for the fluoroquinolones. 213, 2000.
Chromosomal and plasmid-mediated efflux transporter proteins
may be quite specific for antibiotics and metabolic product substrates
and are regulated by numerous genes and gene products. Repressors transformations are uncommon and require unique circumstances
are also present and are highly regulated to prevent the accidental over- involving genes, binding, uptake, and integration.
production of efflux pumps. Tetracyclines can derepress this system, Transduction is the movement of DNA from one bacterium to
leading to an overproduction of efflux proteins and increasing resis- another by a bacteriophage (bacterial virus) intermediary. Conjugation
tance to themselves and any other antibiotics carried by these proteins. is the self-transfer of genetic information by plasmids or transposons
to other microorganisms, generally by physical contact with a sex pilus
Genetic Variations in Bacteria in gram-negative organisms and stimulated by various pheromones
Microorganisms possess three mechanisms for genetic variation: (1) (small peptides).
local nucleotide changes in the genome, (2) rearrangement of genomic A summary or bacterial resistance mechanism by class of antimi-
sequences, and (3) horizontal acquisition of DNA from other microor- crobial drugs is given in Table 33-1.
ganisms (horizontal gene transfer).
Horizontal gene transfer between organisms occurs by three
mechanisms—transformation, transduction, and conjugation—and
ADVERSE REACTIONS TO ANTIBIOTICS
use numerous transposable elements that involve mobile genetic ele- This section discusses adverse drug reactions, some of which are
ments that are passed from one bacterium to another. These mobile unique to antibiotics; others are not exclusive to antimicrobials, but
genetic elements include naked DNA, plasmids, bacteriophages, trans- they are clinically significant for some antimicrobial drugs.
posons, and integrons.
Plasmids are circular, double-stranded DNA outside the chromo- Allergies
somes. Bacteriophages are viruses that infect bacteria and can medi- The penicillins are an example of antibiotics associated with a signifi-
ate genetic transfer. Transposons are DNA segments that cannot cant number of allergies among the population. These can range from
self-­replicate, but they can self-transfer between plasmids, bacterio- a mild rash to acute anaphylaxis.
phages, and chromosomes. Integrons capture and disseminate genes
by site-specific integration of DNA (gene cassettes). Antibiotic Resistance
During transformation, bacteria acquire “naked” DNA from An inevitable effect of the use of antibiotics is the development of resis-
their environment to incorporate into their genome. Such genetic tance on the part of bacteria. On balance, the evidence is substantial
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 461

that antimicrobial agents at any dose or concentration for virtually any fatal pseudomembranous colitis (PMC). Of the 25 million people
length of time do select for resistance and promote the acquisition and affected by serious diarrhea annually in the United States, approx-
transfer of drug-resistant genes. Many of these species exhibit extraor- imately 10% of these cases are the result of antibiotics, particularly
dinary resistance patterns: 50% to 100% of Salmonella, staphylococci, broad-spectrum agents. Most of these cases of antibiotic-associated
and enteric bacilli are resistant to tetracycline, and 32% to 47% are diarrhea are not clinically significant and respond to drug discontinu-
resistant to β-lactams, with 49.7% exhibiting polyantibiotic resistance; ance and rehydration if necessary.
30% of Staphylococcus aureus are resistant to ciprofloxacin and 47% to Any antibiotic is capable of inducing diarrhea, colitis, or PMC,
tetracycline; 72% of Campylobacter in humans and 99% in chickens but the most common agent involved is amoxicillin, followed by
and pigs are resistant to ciprofloxacin; and E. coli exhibits 70% to 94% third-generation cephalosporins and clindamycin. When the colonic
resistance to amoxicillin and 62% to 98% resistance to tetracycline. flora are disturbed by antibiotics or disease, the colonization resistance
Very low (nanogram/nanomolar) concentrations of antibiotics found of the gastrointestinal tract is reduced by the suppression of natural
in the food chain used in nature to control bacterial ecologic niches antagonists of C. difficile such as Bacteroides, Lactobacillus, pseudo-
induce resistance patterns; subtherapeutic dosages in humans are suf- monads, staphylococci, streptococci, peptostreptococci, enterococci,
ficient to challenge microorganisms and lead to resistance. and E. coli.
The mere presence of a β-lactam antibiotic produces a hundred- The fear of inducing a potentially fatal case of PMC has led to a
fold to a thousandfold increase in induction of β-lactamase in micro- reluctance to use clindamycin because early and faulty preliminary
organisms producing extended-spectrum β-lactamases. E. coli carries data reported a 10% association of PMC with the drug. More recent
resistance genes that are not expressed until tetracycline is present. data indicate that incidence of antibiotic-associated diarrhea and
Concentrations of tetracyclines at 0.1 to 1 μg/mL/ per gram in meat CDAC associated with clindamycin in community use of the drug is
cause the dissemination of resistance genes in the human gastrointesti- very low. The overall risk rate for community-acquired C. difficile–
nal tract, and 1 μg/mL of tetracycline in drinking water results in a ten- associated PMC from retrospective data may be 1 per 10,000 antibiotic
fold increase in the transfer of conjugative plasmids from Enterococcus prescriptions, and the risk of hospitalization may be 0.5 to 1 per 100,000
faecalis to Listeria monocytogenes. patient years. The incidence rate was calculated to be 1.6 per 100,000
The use of antibiotics can also promote the transfer of resistance persons exposed to ampicillin, 2.9 per 100,000 persons exposed to
genes from one species to another. In oral plaque biofilm, tetracycline dicloxacillin, and 2.6 per 100,000 persons exposed to tetracycline, with
resistance genes can be transferred from Bacillus subtilis to strepto- no antibiotic-associated diarrhea seen in the 1509 patients receiving
cocci, illustrating that non-oral bacteria have the potential to trans- oral or topical clindamycin.
fer genes to opportunistic oral microorganisms. The self-transfer of Statistically, PMC is more likely to occur with amoxicillin than
Bacteroides conjugative transposons can be increased a hundredfold to clindamycin. Clinicians should refrain from unnecessary antibiotic
a thousandfold by the presence of low levels of tetracycline (1 μg/mL). therapy in patients within the first 2 months after the elimination of
Oral streptococci can harbor tetracycline resistance genes in dental CDAD. Any elective dental procedure requiring antibiotic treatment
plaque and disseminate such genes by mobile elements to other micro- or prophylaxis would best be postponed for this 2-month period. If
flora: Enterococcus faecalis, Veillonella, and other streptococci. Salyers antibiotic therapy is required, the use of antibiotics far less commonly
and colleagues stated that “the fact that tetracycline acts as an inducer associated with CDAD (penicillin V, macrolides) is appropriate.
of transfer gene expression illustrates how the use of an antibiotic An example of superinfection in the mouth is the growth of Candida
could accelerate the spread of antibiotic resistance genes not only by albicans as a result of treatment with an antibiotic, especially one with a
selecting for their acquisition but also by stimulating their transfer.” broad or extended spectrum or metronidazole.
Exposure to antibiotic doses at the low concentrations seen in agri-
culture and aquaculture, as therapy for inflammatory or other diseases, Nephrotoxicity
or for growth effects is a major concern for public health. These antibi- The kidney is sensitive to the effects of some antibiotics, especially
otics can make their way into the food and water supply and alter body aminoglycoside and peptide antibiotics. Combining two drugs with
flora or promote emergence of resistant microbes or the transfer of overlapping toxicity compounds the problem. Mechanisms of renal
resistance genes. Perhaps more importantly, the wide use of antibiotics toxicity involve concentration of the drug in renal tubule cells, leading
in farm animals leads to organisms that are resistant to one or multiple to the lack of ability to concentrate urine and eventual reduced glomer-
antibiotics. ular filtration rate.

Superinfection Ototoxicity
A significant and unappreciated adverse effect of antibiotics is the Aminoglycosides and peptide antibiotics also are associated with dam-
potential to decrease colonization resistance of indigenous anaer- age and other effects on the eighth cranial nerve leading to vestibu-
obic flora in the digestive tract and other anatomic areas (skin, oral lar and cochlear dysfunction. Hearing loss, especially high frequency,
mucosa). The role of colonization resistance is to limit the concentra- occurs and can be irreversible due to the concentration of the drugs in
tion of potentially pathogenic flora of either an exogenous or endog- the endolymph and perilymph and damage to the cochlea.
enous nature in a given body part. Removal of indigenous flora by
antibiotics can promote growth of microorganisms not sensitive to the Antibiotic-Induced Photosensitivity, Photoallergy, and
drug (superinfection). Many superinfections result from a reduction Phototoxicity
in the endogenous microorganisms important for colonization resis- Some antibiotics (along with phenothiazine antipsychotics) are among
tance, with the most notable example being antibiotic-induced diar- the most common drugs inducing skin reactions on exposure to sun-
rhea and colitis. light. Photosensitivity may occur in one of two forms: (1) phototoxicity,
Adverse colonic effects of antibiotics range from simple diarrhea in which chemicals (drugs) are deposited in the skin, absorb ultraviolet
(antibiotic-associated diarrhea) to mucosal inflammatory diarrhea/­ light, and transfer the energy to local tissue, resulting in inflamma-
colitis (antibiotic-associated colitis), with or without associated Clos­ tory responses, or (2) photoallergy, in which sunlight causes a hapten
tridium difficile (C. difficile–associated colitis [CDAC]), to potentially to become a complete antigen in the skin, eliciting an immediate or
462 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

a delayed allergic reaction. The signs and symptoms (erythema, urti- times the half-life of the drug). The oral contraceptive should never
caria, eczema, lichenoid dermatitis, bullous lesions) may be the same, be stopped during antibiotic therapy because it is the most effective
but the mechanisms are different (photoallergy may need a sensitizing means of contraception with the exception of abstinence.
dose unless the drug is continually taken for ≥5 to 10 days). The most The one antibiotic that has been shown to reduce the effect of oral
common antibiotics that induce photosensitivity are sulfonamides, contraceptives is rifampin. In this case the mechanism is an induction
tetracyclines, and fluoroquinolones. Photosensitivity is managed by of hepatic metabolism by rifampin. Women taking an oral contracep-
discontinuing the drug, avoiding sunlight, and wearing protective tive and then prescribed rifampin must use a means of birth control
clothing. other than an oral contraceptive.

Long QT Interval Syndrome


NEW ANTIMICROBIAL APPROACHES
Long QT interval syndrome is a cardiac disorder caused by ion channel
abnormalities that prolong the time interval between the beginning of Pharmaceutical companies see new antibiotic development as prob-
the QRS complex and the end of the T wave on the electrocardiogram lematic for economic, regulatory, and scientific reasons. As a result, 10
(see Chapter 19). Antibiotics that have been implicated in the cause of of the 15 largest drug companies have reduced or eliminated antibiotic
torsades de pointes include fluoroquinolones (gatifloxacin, levofloxa- research since 1999.
cin, moxifloxacin, sparfloxacin), macrolides (erythromycin, clarithro- The scientific difficulty with developing new antibiotics is that all the
mycin), and clindamycin. easy targets in bacteria have already been discovered with possibly only
The Food and Drug Administration (FDA) Adverse Event Reporting a few remaining. Formulating “new” antibiotics that are merely deriv-
System has found that 77% were caused by macrolides and 23% by fluo- atives of existing antibiotics will not solve all the problems of microbial
roquinolones; 89% to 95% were in older patients; 9% to 13% were fatal; resistance. Entirely new approaches to unique mechanisms of antibiotic
the mean time to the adverse event was 4 to 5 days; and 42% to 62% had action attacking heretofore unknown microbial metabolic processes
cardiac disease, 7% to 11% had renal disease, and 17% had low blood require a much better basic understanding of microbial life and consid-
K+ or Mg++ levels. The risk rate has been estimated to be 1 per 1 million erable risk-taking on the part of the pharmaceutical industry.
exposures to ciprofloxacin, 3 per 1 million exposures to clarithromycin,
and 14.5 per 1 million exposures to sparfloxacin (withdrawn from the
ANTIBACTERIAL ANTIMICROBIAL DRUGS
market in the United States).
Antibacterial drugs are primarily classified according to their chemical
Antibiotics and Oral Contraceptives class and mechanism of action. They also can be distinguished based
In response to a few case reports, in the 1980s the FDA issued a warn- on spectrum and adverse effects. In addition to these aspects of anti-
ing that antibiotics may interfere with the action of oral contracep- microbial drugs, the therapeutic uses, including dental applications, of
tives, potentially resulting in unwanted pregnancies. The proposed each class of drugs are discussed.
mechanisms of reduced contraceptive blood concentrations leading
to decreased efficacy include (1) increased urinary/fecal excretion β-Lactam Antibiotics
from antibiotic-induced diarrhea, (2) increased microsomal liver Penicillin belongs to the β-lactam family of antibiotics, which remain
metabolism, (3) receptor displacement, (4) reduced gastrointestinal the most widely used antibiotics in the world. β-lactams are composed
absorption, and (5) reduced enterohepatic circulation. The antibiotic of five different groups of antibiotics, with the β-lactam nucleus as the
rifampin stimulates the liver metabolism of the oral contraceptives, common feature: penicillins, cephalosporins, carbapenems, mono-
reducing blood levels. No other experimental data or controlled clin- bactams, and carbacephems. Penicillins and cephalosporins are the
ical studies have documented the interference of any other antibiotics most important with carbapenems (imipenem, meropenem, ertape-
with the activity of oral contraceptives. nem), monobactams (aztreonam), and carbacephems (loracarbef)
The reasoning is that some antibiotics, especially broad- or extend- reserved for serious infections such as nosocomial (hospital-acquired)
ed-spectrum antibiotics, reduce enteric bacteria that metabolize the infections. β-lactams vary greatly from one another in their spectrum
conjugated forms of the estrogens and progestins that make up the of antimicrobial activity, ranging from an extremely narrow spectrum
contraceptive. As a result, less regenerated form of the hormones (e.g., β-lactamase–resistant penicillins) to a very wide spectrum (e.g.,
would be available for reabsorption from the gut, resulting in less imipenem and some cephalosporins).
effect from the contraceptive. However, several studies document no
effect of antibiotics on the blood levels of ethinyl estradiol, norethin- Penicillins
drone, and progesterone in patients taking doxycycline (100 mg/day Penicillin is a generic term for a group of antibiotics that share the
for 7 days), tetracycline (500 mg every 6 hours for 10 days), and cipro- β-lactam ring nucleus, similar adverse drug reactions, and similar
floxacin (500 mg three times/day for 7 days). No effort has been made mechanism of action, but differ in their antibacterial spectrum, phar-
to determine whether the failure rate of oral contraceptives in women macokinetics, and resistance to β-lactamase enzymes.
taking antibiotics is greater than the normal failure rate of oral con- Classification. Penicillin is a cyclic dipeptide consisting of two
traceptives in women not taking antibiotics. No official authoritative amino acids (d-valine, l-lysine), a particular molecular configuration
body has ever examined this alleged drug interaction to investigate the unknown in higher life forms. The synthesis in 1958 of the basic structure
evidence and make a recommendation. of penicillins (6-aminopenicillanic acid) allowed for its manipulation
From a purely scientific point of view, no reason exists to believe by the addition of various side chains to the β-lactam and thiazolidine
that any antibiotics other than rifampin interfere with the action of rings (Fig. 33-4). Different salts (Na+, K+, procaine, benzathine)
oral contraceptives. From a medicolegal point of view, the dentist may were also created for pharmacokinetic purposes. On the basis of
wish to advise a patient taking oral contraceptives and receiving antibi- these modifications, penicillins can be divided into four groups: (1)
otics to use an additional contraceptive method or practice abstinence penicillin G and penicillin V, (2) antistaphylococcal penicillins that
during the time the antibiotic is present and for several days after its are resistant to β-lactamase produced by staphylococci, (3) amino-
termination to allow for complete antibiotic excretion (usually five penicillins with an extended-spectrum, and (4) extended-spectrum
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 463

penicillins with added activity against gram-negative organisms like and dicloxacillin are examples. (Although similar to penicillin V in
Pseudomonas aeruginosa (Table 33-2). Some penicillins are combined other ways, penicillin G is acid labile and is not often used orally.)
with a separate β-lactamase inhibitor to protect the penicillin from the Penicillinase-resistant penicillins are resistant to some β-lactamases.
enzyme. Bacteria, particularly staphylococci, develop resistance to penicillins
Acid-stable penicillins are resistant to break down in stomach acid, chiefly through the elaboration of β-lactamase enzymes (penicilli-
indicating their usefulness as oral drugs. Penicillin V, amoxicillin, nases) that inactivate the penicillins by cleavage of the 6-aminopeni-
cillanic acid nucleus to yield penicilloic acid derivatives (see Fig. 33-4).
The production of staphylococcal penicillinase is encoded in a plasmid
R Group and may be transferred to other bacteria. Methicillin was the first semi-
synthetic derivative to be introduced that was stable in the presence
O of β-lactamase. Subsequently, nafcillin and three isoxazolyl derivatives
H (oxacillin, cloxacillin, and dicloxacillin) were marketed.
HN S
CH3 Extended-spectrum penicillins are represented by two groups of
CH3
penicillin derivatives. One group is the aminopenicillin group: ampi-
Penicillin G N
O cillin, amoxicillin and bacampicillin. The latter is a drug that is rap-
R Groups
OH idly hydrolyzed in vivo to yield ampicillin. The second group contains
O ticarcillin and piperacillin that exhibit activity against Pseudomonas
Penicillinase and indole-positive Proteus species.
O Mechanism of action. Early in the discovery of penicillin, it was
Penicillin V noted that the drug acted only on rapidly dividing organisms, and it was
Cl O later determined that bacterial cell wall precursors (the Park nucleotides)
N CH3 O accumulated in sensitive bacteria exposed to the penicillins. Penicillin
H
was determined to be a structural analogue of d-alanine; the final step
HN S
CH3 in the formation of the bacterial rigid cell wall was a transpeptidation
Cl HN CH3 reaction involving the enzymatic removal of a terminal d-alanine to
Dicloxacillin O allow for the formation of the cross-linked peptidoglycan cell wall
OH
NH2
Benzylpenicilloic acid OH (Fig. 33-5). β-Lactams are competitive inhibitors of various enzymes
O (transpeptidases, carboxypeptidases), collectively termed penicillin-
sensitive enzymes, or more commonly PBPs. β-lactams promote the
HO formation of cell wall–deficient microorganisms of different shapes
(oval, oblong, spherical) depending on the particular PBP affected,
Amoxicillin
which cannot maintain their internal osmotic pressure and eventually
S burst. The mechanism of action of β-lactams is a classic example of
Ehrlich’s goal of the “magic bullet,” or more specifically a chemical
that inhibits a cellular activity present only in bacteria (a rigid cell wall)
HO O and not found in mammalian cells.
Ticarcillin In some bacterial species, β-lactams have an additional mechanism
FIG 33-4  Structure of penicillin G and structures of penicillin V, dicloxa- of action as they activate an enzyme, muramyl synthetase, responsible
cillin, amoxicillin, and ticarcillin, as shown by replacement of the R group for the separation of daughter cells after cell division. Activation of this
of penicilloic G. Also shown is the effect of penicillinase in producing the enzyme in the absence of cell division produces lysis of the cell wall
penicilloic acid metabolite, which is inactive as an antimicrobial drug. (autolysis) and results in bacterial “suicide.”

TABLE 33-2  Penicillin Groups


Group Examples Acid Stable Spectrum Penicillinase Resistant
Penicillin G and congener Penicillin G No Narrow No
Penicillin V Yes Narrow No
Anti-staphylococcal Methicillin* No Narrow Yes‡
Nafcillin Somewhat|| Narrow† Yes‡
Oxacillin Yes Narrow† Yes‡
Cloxacillin Yes Narrow† Yes‡
Dicloxacillin Yes Narrow† Yes‡
Amino-penicillins, extended-spectrum Ampicillin Yes Extended No
Amoxicillin Yes Extended No
Extended-spectrum§ Ticarcillin* No Extended§ No
Piperacillin No Extended§ No
*No longer used clinically.
†Limited to use against staphylococcal organisms.
‡Resistant to penicillinase from Staphylococcus aureus.
§Spectrum includes other gram-negative bacteria (e.g., Pseudomonas aeruginosa and indole-positive Proteus).
||Although nafcillin is somewhat acid stable, it is used parenterally.
464 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

FIG 33-5  Bacterial cell wall synthesis. Details are discussed in the text. The sites of action of various drugs are
shown. The lipid carrier that is inhibited by bacitracin is shown as a wavy line. (From Cohen J, et al.: Infectious
diseases, ed 3, St Louis, 2010, Mosby.)

Considering these mechanisms, it is apparent why consistently penicillins have an extended spectrum and greater activity against
high blood levels of β-lactams are required for optimal success (not some gram-negative bacilli.
all bacteria divide at the same time) and why penicillins do not kill Penicillin G or penicillin V are drugs of choice against the VGS, S.
rapidly (it takes time for enzyme inhibition and eventual microor- pneumoniae, S. Pyogenes, Peptostreptococcus, E. faecalis, Fusobacterium
ganism rupture). This realization that β-lactams kill slowly has raised nucleatum, Actinomyces israelii, Clostridium tetani, Clostridium per­
questions about the mechanism of action in endocarditis prophy- fringens, Leptotrichia buccalis, N. meningitidis, and non-β-lactamase–
laxis: whether they act only (or at all) by microbial killing or rather producing Prevotella and Porphyromonas. Amoxicillin alone or with
by cell wall alteration to retard attachment of the bacteria to damaged clavulanate has activity against the same organisms but also Eikenella
cardiac valves. corrodens, K. pneumoniae, Enterobacter, Moraxella (Branhamella)
Antibacterial spectrum. Penicillin G and penicillin V are narrow- catarrhalis, Bacteroides fragilis, non–methicillin-resistant and (with
spectrum antibiotics, showing activity against mostly gram-positive clavulanate) β-lactamase-producing Prevotella and Porphyromonas E.
cocci and gram-positive bacilli and gram-negative cocci. Other coli, Haemophilus influenzae, H. pylori, and P. mirabilis (Table 33-3).
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 465

TABLE 33-3  Penicillins as Drugs of Choice TABLE 33-4  Disease Entities for Which
or Alternative Agents (Penicillin G, Penicillin Penicillin G, Penicillin V, and Amoxicillin Are
V, Ampicillin, or Amoxicillin Unless Otherwise of Major Use
Indicated) Abscesses, including orodental
Acinetobacter* Bacteremia (gram-positive)
Actinomyces israelii Endocarditis
Bacillus anthracis Gas gangrene
Bacteroides* Mastoiditis
Campylobacter fetus* Meningitis
Capnocytophaga canimorsus Orodental infections
Citrobacter freundii* Osteomyelitis
Clostridium perfringens Pericarditis
Clostridium tetani Periodontal infections
Eikenella corrodens Pharyngitis
Enterobacter* Pneumonia
Erysipelothrix rhusiopathiae Rat-bite fever
Fusobacterium nucleatum Scarlet fever
Group A, B, C, and G streptococci Suppurative arthritis
Listeria monocytogenes Syphilis
Neisseria meningitidis Vincent’s stomatitis
Pasteurella multocida Weil’s disease
Peptostreptococcus micros Wound infections
Serratia marcescens*
These diseases are caused by various gram-positive cocci and bacilli
Proteus mirabilis and some gram-negative organisms, spirochetes, and anaerobic
Spirillum minus microorganisms. Susceptibility testing may be essential for some to
Streptobacillus moniliformis determine therapeutic mean inhibitory concentrations.
Staphylococcus aureus/Staphylococcus epidermidis†
Streptococcus bovis penicillin (Fig. 33-6). The route of excretion is primarily by the kid-
Treponema pallidum neys, with limited liver metabolism. Rapid kidney excretion accounts
VGS for the short half-lives of most penicillins, 90% of which is due to
VGS, Viridans group streptococci. active proximal tubular secretion, while the remaining 10% is due to
*Imipenem/meropenem. glomerular filtration. This is the basis for occasionally extending the
†β-Lactamase-resistant penicillins if methicillin susceptible. half-lives of some penicillins by combining the penicillin with probe-
From Handbook of antimicrobial therapy. Choice of antibacterial drugs. necid, which inhibits active tubular transport.
Med Lett Drugs Ther 20:69-88, 2015; Facts and comparisons, St Louis, The distribution of the penicillins includes most body fluids but
2015, Facts and Comparisons. not the humors of the eye or the brain. Penicillin does not pass through
the blood–brain barrier unless the meninges are inflamed, such as in
Amoxicillin and penicillin V are the initial drugs of choice in oro- meningitis.
facial infections in nonallergic patients, but they are ineffective against β-lactam antibiotics produce time-dependent killing of bacte-
streptococci (VGS) with altered PBPs. The clinical impact of antibiotic ria, and frequent dosing is required to maintain relatively constant
failures against these resistant streptococci and gram-negative β-lact- blood levels with as little fluctuation as possible. The killing power of
amase–producing oral anaerobes is likely to be significant but has yet β-lactams is maximum at three to four times the minimum inhibi-
to be determined by clinical studies. On the basis of the antimicrobial tory concentration (MIC) of susceptible microorganisms. The prime
spectrum of penicillin G and V and other clinical characteristics, the determinant of the efficacy of β-lactams is the length of time the con-
drugs are useful in the treatment of numerous diseases (Table 33-4). centration of the drug in the infected area is greater than the MIC of
Bacterial resistance. Bacteria evade the killing effects of β-lactams the infecting organism.
by three mechanisms: reduced drug binding to PBPs (altered target To be maximally effective, the serum and tissue concentrations
sites), hydrolysis by β-lactamase enzymes (enzymatic inactivation), of β-lactams should be greater than the MIC for 50% to 70% of the
or development of tolerance by the loss of the autolysis mechanism dosing interval. The current package insert recommends dosing inter-
(penicillin becomes bacteriostatic instead of bactericidal). In most vals of 6 hours for penicillin V. Penicillin V and penicillin G have an
species, the principal mechanism is β-lactamase production. elimination half-life of approximately 30 minutes, and consequently,
Absorption, fate, and excretion. Penicillin G (benzylpenicillin) 6-hour dosing intervals may result in very low serum levels in the last
is rarely used orally because of its poor gastric absorption rate. 2 or 3 hours. Continuous intravenous penicillin is receiving greater
Penicillin V and amoxicillin are well absorbed orally, with amoxicillin attention as a way to circumvent this problem.
considerably longer in its half-life (∼1 hour vs ∼30 minutes). Better oral β-lactamase inhibitors. Currently, four agents are available to
absorption argues for the use of amoxicillin over penicillin V, but both bind irreversibly to the catalytic site of susceptible β-lactamases to
drugs are effective in microorganism-sensitive orofacial infections and prevent hydrolysis of β-lactam antibiotics: clavulanic acid, sulbactam,
are equally inactive against VGS with altered PBPs. tazobactam, and avibactam. Clavulanic acid is derived from
Procaine penicillin G and benzathine penicillin G are repository Streptomyces clavuligerus, sulbactam is a semisynthetic penicillinate
forms prepared for intramuscular injection with slow release from the sulfone, tazobactam is chemically related to sulbactam, and avibactam
injection site. These preparations extend the plasma level profiles of is a non-β-lactam inhibitor with broader activity against β-lactamases.
466 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

Aqueous crystalline penicillin G


7 300,000 units

Plasma concentration (units/mL)


Procaine penicillin G
6 300,000 units

Benzathine penicillin G
5 1,200,000 units

0.4
3
0.3
2
0.2

1
0.1

2 3 1 6 9 12 29 1 15 30
Hours after IM injection Days after IM injection
(compressed scale)
FIG 33-6  Comparative plasma concentrations of penicillin G obtained from soluble versus repository intra-
muscular (IM) dosage forms.

All β-lactamase inhibitors have the same mechanism of action, which infections are associated with gram-positive and gram-negative aero-
is to bind to the active site of β-lactamases, where they are converted bic and anaerobic microorganisms, for which an antimicrobial agent
to an inactive product by β-lactamase (“suicide inhibition”). Only with a more extended antibacterial spectrum, such as amoxicillin or
clavulanic acid is orally absorbed. Clavulanic acid is combined with more commonly a β-lactam/β-lactamase agent combined with metro-
amoxicillin, sulbactam with ampicillin, tazobactam with piperacillin, nidazole, may be the agent of choice.
and ceftazidime (see later) with avibactam. Adverse effects. The adverse effects of penicillins are allergic and
The sole therapeutic use of β-lactamase inhibitors is to prevent the nonallergic in nature.
hydrolysis of β-lactam antibiotics in the management of β-­lactamase– Allergic reactions to penicillins are common, while allergic fatal-
producing microorganisms responsible for otitis media and sinusitis ities are far less common. Allergy to penicillins ranges from 0.7% to
(S. pneumoniae, H. influenzae, M. catarrhalis), nosocomial pneumo- 8% in various studies, with a 0.7% to 4% chance of an allergic reaction
nia (methicillin-sensitive S. aureus (MSSA) or K. pneumoniae), intra-­ (average of 2%) during any given course of penicillin therapy. Most
abdominal abscesses from β-lactamase–producing anaerobes and allergic manifestations are maculopapular or urticarial skin reactions.
other microorganisms, and some upper respiratory tract infections. Penicillin may be the most common cause of anaphylactic death
β-Lactamase inhibitor combinations offer no advantage against non-β- in the United States, accounting for 75% of all cases and 400 to 800
lactamase–­producing microorganisms and are ineffective against methi- annual deaths. Estimates of severe penicillin anaphylaxis range from
cillin-resistant S. aureus (MRSA), many coagulase negative staphylococci 0.004% to 0.015% of individuals exposed and, from the point of view
(CoNS) and enterococci, and the inducible β-lactamases produced by of number of exposures, possibly 1 in 1200 to 1 in 2500 penicillin
P. aeruginosa, Serratia marcescens, Enterobacter cloacae, C. freundii, and exposures.
Morganella morganii. These β-­lactamase inhibitor combinations can Eventually, 1% to 10% of the general population exposed to thera-
often be useful as alternative antibiotics against Bacteroides, M. catarrha­ peutic penicillin has an allergic reaction, with a higher positive history
lis, E. coli, K. pneumoniae, indole-positive Proteus and others (see Tables with increased age. The incidence of allergy varies with the route of
33-3 and 33-5). administration: oral (0.3%), intravenous (2.5%), and intramuscu-
Therapeutic uses in dentistry. Because the oral route is the safest, lar (5%); the lower incidence by the oral route has been questioned
most convenient, and least expensive mode of drug administration, because of limited data.
it is favored in the treatment of dental patients. Currently, penicillin It is probable that an acute penicillin-allergic reaction is less com-
V and amoxicillin are the most frequently prescribed antibiotics for mon in children and elderly patients, but fatal reactions may be more
chemotherapy of infections of dental origin. likely in elderly patients because of their compromised cardiopulmo-
In some instances, penicillins G and V and amoxicillin are unsuit- nary function. Whether certain individuals are predisposed to peni-
able for treating oral infections. Some dental infections are caused by cillin allergy remains unsettled. Risk factors for penicillin allergies
β-lactamase (penicillinase)–producing organisms, and in such cases include multiple allergies to other drugs, particularly other antibiot-
the appropriate antibiotic is a penicillinase-resistant penicillin deriv- ics (“multiple allergy syndrome”), or atopic disease (asthma, allergic
ative, erythromycin, or clindamycin. Patients who have been receiv- rhinitis, nasal polyps). Cross-allergenicity exists for all penicillins and,
ing extended prophylactic therapy with penicillin for the prevention even to a certain extent, other β-lactam antibiotics.
of rheumatic fever generally require another antibiotic if they acquire In individuals with a positive history of penicillin allergy, 15% to
an infection or require endocarditis prophylaxis. Certain periodontal 40% exhibit allergy on re-exposure to penicillin, and individuals with
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 467

TABLE 33-5  Drugs Used to Treat Infections Caused by Specific Microorganisms


Microorganism Drug of First Choice Alternative Drugs*
Gram-Positive Cocci
Staphylococcus species
Methicillin-sensitive Penicillinase-resistant penicillin A cephalosporin, vancomycin, imipenem, meropenem, clindamycin, a
(e.g., dicloxacillin) fluoroquinolone, linezolid, tedizolid, daptomycin, telavancin
Methicillin-resistant Vancomycin ± rifampin Daptomycin, quinupristin/dalfopristin, linezolid, a fluoroquinolone,
doxycycline, trimethoprim-sulfamethoxazole, telavancin, dalbavancin,
oritavancin
Streptococcus pyogenes Penicillin G or V A cephalosporin, erythromycin, clindamycin, vancomycin, clarithro-
mycin, azithromycin, linezolid, tedizolid, daptomycin, telavancin,
dalbavancin, oritavancin
Streptococcus viridans group
Oral infections Penicillin G or V Erythromycin, clindamycin, a cephalosporin
Bacteremia or endocarditis Penicillin G with or without gentamicin Ceftriaxone, vancomycin
Streptococci, anaerobic (Peptost- Penicillin G or V A cephalosporin, clindamycin, vancomycin, daptomycin, telavancin
reptococcus)
Streptococcus ­pneumoniae Penicillin G or V, amoxicillin A cephalosporin, trimethoprim/sulfamethoxazole, erythromycin,
clarithromycin, azithromycin, clindamycin, levofloxacin, gemifloxa-
cin, moxifloxacin, meropenem, imipenem, ertapenem, doripenem,
vancomycin, telavancin, a tetracycline
Enterococcus faecalis Ampicillin, amoxicillin, penicillin Vancomycin with gentamicin, daptomycin ± gentamicin, linezolid, ampi-
G + gentamicin or ceftriaxone cillin with imipenem (nitrofurantoin or fosfomycin, urinary tract only)
Enterococcus faecium (vancomycin Daptomycin (+gentamicin or ampicillin) Ampicillin + gentamicin, linezolid in combination with other active
resistant) drug, quinupristin/dalfopristin ± ampicillin (nitrofurantoin or fosfomy-
cin, urinary tract only)
Gram-Negative Cocci
Neisseria gonorrhoeae Ceftriaxone Ceftizoxime, cefoxitin + probenecid, cefixime, cefpodoxime
Neisseria meningitidis Ceftriaxone, cefotaxime, penicillin G A fluoroquinolone, meropenem, chloramphenicol
Moraxella (Branhamella) A fluoroquinolone, cefuroxime Trimethoprim-sulfamethoxazole, amoxicillin/clavulanate, erythromycin,
catarrhalis clarithromycin, azithromycin, doxycycline, cefotaxime, ceftizoxime,
ceftriaxone, cefpodoxime
Gram-Positive Bacilli
Bacillus anthracis Ciprofloxacin, doxycycline Penicillin G, amoxicillin, erythromycin, imipenem, clindamycin, levofloxacin
Clostridium difficile Metronidazole, vancomycin Fidaxomicin
Clostridium perfringens Penicillin G Clindamycin, imipenem, meropenem, ertapenem, doripenem, metroni-
dazole, chloramphenicol
Clostridium tetani Metronidazole Penicillin G, doxycycline
Corynebacterium ­diphtheriae Erythromycin Penicillin G
Corynebacterium jeikeium Vancomycin Daptomycin
Gram-Negative Bacilli
Bacteroides (oropharyngeal Penicillin G Cefotetan, cefoxitin, clindamycin, metronidazole, ampicillin-sulbactam,
strains) amoxicillin-clavulanate
Capnocytophaga ­canimorsus Amoxicillin/clavulanic acid Cefotaxime, ceftriaxone, ceftizoxime, clindamycin, a fluoroquinolone,
imipenem, meropenem, vancomycin, doxycycline
Eikenella corrodens Ampicillin ± sulbactam, amoxicillin ± clavulanate Azithromycin, clarithromycin, ceftriaxone, doxycycline,
Escherichia coli Cefotaxime, ceftriaxone, ceftazidime, cefepime Ampicillin ± (gentamicin, tobramycin, or amikacin), aztreonam, ampicil-
lin/sulbactam, amoxicillin/clavulanic acid, piperacillin/tazobactam,
trimethoprim/sulfamethoxazole, imipenem, meropenem, doripenem,
ertapenem, a fluoroquinolone, tigecycline
Fusobacterium species Penicillin G, penicillin V, metronidazole Clindamycin, cefoxitin, imipenem, meropenem
Haemophilus influenzae Cefotaxime, ceftriaxone, trimethoprim- Cefaclor, cefuroxime, cefotaxime, cefpodoxime, ceftizoxime, ceftri-
sulfamethoxazole axone, cefixime, a fluoroquinolone, doxycycline, clarithromycin,
azithromycin, ampicillin or amoxicillin ± penicillinase inhibitor
Klebsiella pneumoniae Cefotaxime, ceftriaxone, cefepime An aminoglycoside, aztreonam, a fluoroquinolone, imipenem,
meropenem, ertapenem, doripenem, amoxicillin/clavulanic acid,
ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavula-
nate, tigecycline, trimethoprim/sulfamethoxazole
Continued
468 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

TABLE 33-5  Drugs Used to Treat Infections Caused by Specific Microorganisms—cont’d


Microorganism Drug of First Choice Alternative Drugs*
Legionella pneumophila Azithromycin or ciprofloxacin or other fluoroquinolone Erythromycin, doxycycline
Leptotrichia buccalis Penicillin G, penicillin V Clindamycin, doxycycline, erythromycin
Proteus mirabilis Ampicillin An aminoglycoside, a cephalosporin, a fluoroquinolone, ticarcillin/­
clavulanate, piperacillin/tazobactam, aztreonam, imipenem, mero-
penem, ertapenem, doripenem, trimethoprim-sulfamethoxazole
Pseudomonas aeruginosa Ticarcillin/clavulanate or piperacillin/ (Aztreonam, ceftazidime, cefepime, meropenem, doripenem, or
tazobactam ± an aminoglycoside, ciprofloxacin ­imipenem) + an aminoglycoside, levofloxacin
Salmonella typhi Ceftriaxone, ciprofloxacin, levofloxacin Amoxicillin, ampicillin, azithromycin, trimethoprim-sulfamethoxazole,
chloramphenicol
Shigella species A fluoroquinolone Trimethoprim-sulfamethoxazole, azithromycin, ceftriaxone
Other Microorganisms
Mycobacterium ­tuberculosis Isoniazid + rifampin + pyrazinamide ± ethambutol or Amikacin, kanamycin, cycloserine, moxifloxacin, levofloxacin,
streptomycin ­ciprofloxacin, ofloxacin, capreomycin, kanamycin, ethionamide,
para-aminosalicylic acid, bedaquiline (all in combinations)
Actinomyces israelii Penicillin G, penicillin V Doxycycline, erythromycin, clindamycin
Nocardia asteroides Trimethoprim/sulfamethoxazole ± amikacin +  A tetracycline, sulfisoxazole, amikacin, imipenem, meropenem,
imipenem or meropenem ­ceftriaxone, linezolid, cycloserine
Treponema pallidum Penicillin G Ceftriaxone, doxycycline
Chlamydia psittaci A tetracycline Azithromycin, a fluoroquinolone
Rickettsiae Doxycycline Azithromycin, clarithromycin, chloramphenicol
Candida albicans†
Oral lesions Clotrimazole, nystatin, miconazole Itraconazole, fluconazole, posaconazole, caspofungin (all for more
serious infections)
Systemic infections Fluconazole, itraconazole, caspofungin, anidulafungin, Posaconazole, amphotericin B
micafungin
Viruses†
Herpes simplex
Orolabial Acyclovir, famciclovir, valacyclovir Penciclovir, docosanol
Keratitis Trifluridine, ganciclovir
Genital infection Acyclovir, famciclovir, valacyclovir
Encephalitis Acyclovir
Human immunodeficiency virus Non-nucleoside reverse transcriptase
(preferred regimens) inhibitor-based regimen
Efavirenz + tenofovir disoproxil fumarate + emtricitabine
Protease inhibitor-based regimen
Atazanavir or darunavir + ritonavir + abacavir + 
tenofovir disoproxil fumarate + emtricitabine
Integrase strand transfer inhibitor-based
regimen
1. Raltegravir or elvitegravir/cobicistat + tenofovir
disoproxil fumarate + emtricitabine
2. Dolutegravir + either abacavir + lamivudine or
­tenofovir DF + emtricitabine
Influenza A and B Oseltamivir, zanamivir
*Listing does not include all alternative drugs.
†See Chapter 34 for discussion of antifungal and antiviral drugs.

Adapted from the following: Treatment guidelines: Antiviral drugs, The Medical Letter 127:19-30, 2013; Abramowicz M, Zuccotti G, editors: Hand-
book of antimicrobial drugs: drugs for bacterial infections, The Medical Letter ed 20, 2015, New Rochelle, NY;
Treatment guidelines: drugs for HIV infection, The Medical Letter vol. 12, issue 138:7-16, 2014.

a positive history of penicillin allergy have a four to six times greater to penicillin and then have an allergic reaction to the drug on the next
likelihood of a subsequent reaction than individuals with a negative (third) exposure by resensitization.
history. The serum half-life of penicillin IgE antibodies ranges from 10
to more than 1000 days; the risk of recurrent penicillin allergy is higher Allergy Testing
in individuals with antibodies with long half-lives or repeated peni- Because IgE antibody levels to penicillin are variable, skin testing for
cillin exposures. Few data are available regarding whether the 60% to penicillin allergy becomes problematic. The incidence of positive skin
85% not exhibiting allergy on re-exposure reacquire the IgE antibodies tests in individuals with a history of penicillin allergy ranges from 4%
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 469

to 91% depending on the accuracy of the patient history, the haptens (INR) in patients taking oral anticoagulants either through impaired
in the test solution, and the time elapsed between the allergic reaction platelet function or altered gastrointestinal microbial flora. Untoward
and the skin test. bleeding may also occur, even in patients not taking coumarin anti-
Penicillin skin testing can be considerably valuable in determining coagulants, and is dose-dependent, with a maximum effect 3 to 7 days
who might have a severe anaphylactic reaction. Approximately 95% after penicillin is begun, with a return to a normal bleeding time in 72
of penicillin-allergic individuals form the penicilloyl-protein conju- to 96 hours; this bleeding has been reported after a dental extraction.
gate (the major antigenic determinant), and approximately 5% form The mechanism is likely due to an altered adenosine 5’-diphosphate–
the 6-aminopenicillanic acid and other minor antigenic determinants. mediated platelet aggregation response and is seen most commonly in
Penicillin skin tests with the major and minor antigenic determinants patients with underlying chronic illnesses associated with hypoalbu-
eliciting a negative skin test virtually eliminate the risk for a serious minemia and uremia.
IgE-mediated reaction. A positive skin reaction to the minor determi- Drug interactions. Oral penicillins (penicillin G, penicillin
nant mixture indicates a high risk for anaphylaxis. V, amoxicillin) may be antagonized by bacteriostatic antibiotics
Penicillins are primarily associated with IgE-mediated (Gell and (tetracycline, erythromycin, clindamycin). NSAIDs and probenecid
Coombs type I) allergic reactions, but they may also induce cytotoxic may increase the serum half-lives of penicillins by decreasing their renal
(type II) or immune complex (type III) reactions. Type I signs and excretion. Individuals taking β-adrenergic blocking drugs, especially
symptoms include skin erythema, itching, angioedema, urticaria, nonselective ones, may cause a diminished or nonexistent response to
wheezing, hypotension, and bronchospasm resulting from mast cell/ a β-adrenergic receptor agonist given for the treatment of penicillin-
basophil release of histamine along with other tissue allergic mediators. induced anaphylactic bronchospasm. Ampicillin or amoxicillin, when
Type II reactions are caused by circulating IgM or IgG antibodies that taken with allopurinol, can be associated with a non-urticarial rash.
attach to blood cells and induce blood dyscrasias, including hemolytic Contraindications. Penicillins are generally contraindicated in
anemia, leukopenia, thrombocytopenia, and aplastic anemia. Type III individuals allergic to the drugs, but it is well documented that some
reactions result from the deposition of soluble immune complexes on individuals with a previous allergic history may subsequently tolerate
blood vessels and basement membranes resulting in serum sickness, penicillins without allergic manifestations. The best policy is to refrain
vasculitis, and glomerulonephritis. Type IV, delayed-type hypersensi- if possible from penicillin administration to anyone with a positive
tivity reactions are mediated by T lymphocytes. (See figure 3-4.) history. Penicillins may be contraindicated in some individuals taking
Allergic reactions to penicillins can also be classified according to coumarin anticoagulants because untoward bleeding may occur, but
their time of onset. Immediate IgE reactions begin within seconds to this seems to be highly unpredictable and rare in occurrence. Avoid the
1 hour after drug exposure and are the most life-threatening (it is an concurrent use of amoxicillin or ampicillin with allopurinol.
allergy truism that the more rapid the onset of the allergic reaction,
the more serious the consequences). Accelerated reactions begin 1 to Cephalosporins
72 hours after antigen exposure and usually manifest as urticaria or The isolation of the fungus Cephalosporium acremonium (now
angioedema. Late reactions occur after 72 hours and are characterized Acremonium chrysogenum) in 1948 by Brotzu from the harbor sew-
by type II and type IV (eczema-like) Gell and Coombs reactions. Of all age of Sardinia and the subsequent isolation of the active nucleus
fatal anaphylactic reactions, 96% occur within the first 60 minutes after of cephalosporin C (7-amino-cephalosporinic acid) by Florey and
penicillin exposure. Abraham at Oxford University contributed in large measure to a
Other adverse reactions to penicillins are likely to be autoimmune golden age in antimicrobial chemotherapy. The widespread use of
in origin and have an obscure etiology, including maculopapular cephalosporins because of their broad antibacterial spectra and low
rashes, eosinophilia, Stevens-Johnson syndrome, and exfoliative der- toxicity and allergenicity has resulted in widespread microbial resis-
matitis. A maculopapular rash is seen in 2% to 3% of patients late in tance to these agents.
penicillin therapy. Chemistry and classification (Table 33-6). Cephalosporins are
Nonallergic adverse effects. Piperacillin may cause abnormal closely related to penicillins, with a six-membered dihydrothiazine
coagulation times and may produce abnormal liver function test ring replacing the five-membered thiazolidine ring of penicillin
results. Large intravenous doses of penicillins, especially in patients (Fig. 33-7). Both contain the β-lactam ring, as do the monobactams
with compromised renal function, may induce hyperexcitability, sei- and carbapenems discussed later. Side chain modification of the 7-APA
zures, and hallucinations. This is due to sufficient drug getting across nucleus has led to differences in antibacterial spectrum, pharmacoki-
the blood–brain barrier under these conditions. Amoxicillin is the netics, susceptibility to various β-lactamase, affinity for different PBPs,
most common cause of antibiotic-induced diarrhea/colitis because of and occasionally adverse reactions.
its spectrum and widespread use. Penicillins are FDA pregnancy cate- Cephalosporins are most commonly classified according to their
gory B drugs. “generations”: first generation (introduced in the 1960s), second gen-
Approximately 5% to 10% of individuals receiving ampicillin or eration (introduced in the 1970s), third generation (introduced in the
amoxicillin may have a mild pruritic rash, usually beginning on the 1980s), fourth generation (cefepime introduced in 1997), and fifth
trunk and extending to the face, extremities, and extensor portions of the generation (advanced generation) (ceftaroline, introduced in 2010).
knees and elbows. This nonallergic “ampicillin/amoxicillin rash” is not Cephalosporins evolved from early agents primarily active against
associated with antibody formation and is of unknown cause. It does not gram-positive microorganisms (first generation) to agents with a greater
seem to increase the risk of true penicillin allergy. The rash may begin gram-negative spectrum (second generation) to agents with greater
24 hours to 28 days after the drug is begun and may last 90 minutes to activity against various nosocomial pathogens, including P. aeruginosa,
7 days. The incidence of ampicillin/amoxicillin rash is 95% to 100% in B. fragilis, and organisms producing extended-­spectrum and ampi-
individuals with cytomegalovirus infection/mononucleosis and 22% in cillin C (ampC) β-lactamases. The broad spectrum of cephalosporins
individuals given ampicillin or amoxicillin with allopurinol. and their wide use have been major factors in microbial resistance to
Rare and reversible disorders reportedly associated with penicillins these agents. Technically, second-generation agents include true ceph-
include acute pancreatitis, neutropenia, aseptic meningitis, hepatotox- alosporins and cephamycins (cefoxitin, cefotetan, cefmetazole), which
icity, and increased prothrombin time/international normalized ratio are derived from Streptomyces rather than Cephalosporium.
470 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

TABLE 33-6  Classification and Indications of Cephalosporins by Generations and Other β-Lac-
tam Antibiotics
Cephalosporins Major Indications
First Generation
Cefadroxil (Duricef)* Gram-positive cocci, (but not enterococci or methicillin-resistant Staphylococcus aureus)
Cefazolin (Ancef, Kefzol, Zolicef)† Klebsiella pneumoniae, Proteus mirabilis, Escherichia coli, dental prophylaxis (cephalexin,
Cephalexin (Biocef, Keflex, Keftab)* cefazolin)
Cephalothin (Keflin)†
Cephapirin (Cefadyl)†
Cephradine (Velosef)‡
Second Generation
Cefaclor (Ceclor)* Greater effect against some gram-negative organisms than first-generation drugs; cefotetan
Cefamandole (Mandol)† and cefoxitin have activity against anaerobes
Cefonicid (Monocid)†
Cefotetan (Cefotan)†
Cefoxitin (Mefoxin)†
Cefprozil (Cefzil)*
Cefuroxime (Ceftin, Kefurox, Zinacef)‡
Loracarbef (Lorabid)*
Third Generation
Cefdinir (Omnicef)* Penicillin-resistant S. pneumoniae, VGS, multidrug-resistant S. pneumoniae, ­enterococci, and
Cefixime (Suprax)* some β-lactamase–producing organisms
Cefoperazone (Cefobid)† Ceftazidime/avibactam is effective against Enterobacteriaceae, Pseudomonas ­aeruginosa,
Cefotaxime (Claforan)† and organisms producing extended-spectrum β-lactamases
Cefpodoxime (Vantin)*
Ceftazidime (Ceptaz, Fortaz, Tazicef, Tazidime)†
Ceftazidime/avibactam (Avycaz)†
Ceftibuten (Cedax)*
Cefditoren (Spectracef)*
Ceftizoxime (Cefizox)†
Ceftriaxone (Rocephin)†
Fourth Generation
Cefepime (Maxipime)† Gram-negative bacteria, cefepime is more resistant to many β-lactamases
Fifth Generation
Ceftolozane/tazobactam (Zerbaxa) Similar to third generation drugs, but extended-spectrum β-lactamases; effective vs
Ceftaroline (Teflaro)† Enterobacteriaceae, P. aeruginosa, other gram-negative bacteria and methicillin-
resistant S. aureus
Other Lactam Antibiotics
 Carbapenems
Imipenem (with cilastatin in Primaxin)† Highly resistant gram-negative bacilli, including some anaerobes, P. aeruginosa, Campylobacter
Meropenem (Meronem)† fetus, Citrobacter freundii, Enterobacter, Acinetobacter, Serratia marcescens, and Rhodococ-
Ertapenem (Invanz)† cus equi, MSSA, non-penicillin-resistant S. pneumoniae, Bacillus subtilis, Bacillus cereus,
Doripenem (Doribax)† Clostridium perfringens, Bacteroides, E. coli, K. pneumoniae, P. mirabilis, indole-positive
Proteus, Providencia stuartii, Capnocytophaga canimorsus, Haemophilus influenzae
Monobactams
Aztreonam (Azactam)† Aerobic gram-negative bacteria, Enterobacteriaceae, K. pneumoniae, P. mirabilis, C. freundii,
Yersinia enterocolitica, Pasteurella multocida, Salmonella, Shigella, Providencia, Neisseria,
Haemophilus, and P. aeruginosa
*Oral.
†Parenteral.
‡Oral and parenteral.

Adapted from Asbel LE, Levison M, Cephalosporins, carbapenems, and monobactams, Infect Dis Clin N Am 14:435-447, 2000; Facts and compar-
isons, St Louis, 2002, Facts and Comparisons; Karchmer AW, Cephalosporins, In Mandell GL, Bennett JF, Dolin R, editors: Principles and practice
of infectious diseases, ed 5, New York, 2000, Churchill Livingstone; Marshall WF, Blair JE, The cephalosporins, Mayo Clin Proc 74:187-195,
1999; Abramowicz M, Zuccotti G, editors: Handbook of antimicrobial drugs: drugs for bacterial infections, The Medical Letter ed 20, 2015,
New Rochelle, NY.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 471

O General therapeutic uses. Cephalosporins have wide applications


H in the treatment of infections. The utility of these drugs depends on the
HN S generation. Table 33-6 lists indications for the cephalosporins. First-
generation drugs are used to treat infections caused by staphylococci
NH2 N
CH3
and streptococci. They also are useful in surgical and endocarditis
O prophylaxis. Some gram-negative bacilli, such as P. mirabilis and K.
HO O pneumoniae, may be sensitive. A subset of second generation drugs
Cephalexin represented by cefoxitin has good activity against many gram-negative
HO
O
anaerobes.
O
Third-generation drugs have become prominent in the treatment
N
of serious gram-negative coccal and bacillary infections. They are
S very useful in treating meningitis, pneumonia, gonorrhea, and sepsis
H3C
from sensitive organisms. Cephalosporins are often given with ami-
H H noglycosides for gram-negative bacilli infections. There are significant
HN
HO individual differences between members of the third-generation drugs,
NH and not all indications apply to each member. Cefepime is resistant
Imipenem
to many β-lactamases and is effective in treating some gram-nega-
H 2N S tive bacilli that produce β-lactamases. Ceftaroline has activity against
methicillin-resistant staphylococci as well as several gram-negative
N O CH3
O
organisms (see Table 33-6).
Therapeutic uses in dentistry. Cephalosporins have good
N HN N S OH
activity against many orofacial pathogens but limited activity against
O
H 3C O oral anaerobes. These β-lactam antibiotics are also time-dependent
O agents without significant post-antibiotic effects, and the serum and
H3C OH
tissue concentrations of cephalosporins should remain greater than
O the organism’s MIC for at least 60% of the dosing interval to retard
Aztreonam organism regrowth as much as possible. Presently, some cephalosporins
FIG 33-7 Structural formulas for cephalexin, imipenem, and the are choices for prophylaxis during dental procedures; however, these
monobactam aztreonam. Notice the lack of a second ring fused to the drugs are not indicated in acute dental infections unless culture and
­β-lactam ring in aztreonam. sensitivity testing indicate otherwise.
Adverse effects. Serious adverse reactions associated with
cephalosporins are rare, with the major concern being the potential
Mechanism of action. Cephalosporins possess a mechanism for cross-allergy with penicillins. Less common adverse reactions
of action identical to penicillins: inhibition of bacterial cell wall include transient increases in liver enzymes, nephrotoxicity, reversible
peptidoglycan synthesis by inhibition of penicillin-sensitive enzymes neutropenia, eosinophilia and thrombocytopenia, aseptic meningitis,
(transpeptidases, carboxypeptidases) that are responsible for the and disulfiram-like reactions associated with cephalosporins with the
final three-dimensional structure of the rigid bacterial cell wall. methylthiotetrazole side chain (e.g., cefotetan).
Each bacterial species may have different PBPs, and the affinity of The inherent allergic potential of cephalosporins along with their
cephalosporins for these PBPs can vary greatly. Most cephalosporins cross-allergenicity with penicillins is of major concern. Cutaneous
bind to PBP1 and PBP3 of gram-negative organisms, and depending allergic reactions to cephalosporins (rash, pruritus, urticaria) are com-
on which PBPs are inhibited, the resulting bacterial cells may take monly reported to occur in 1% to 3% of patients. Serum sickness or a
different shapes: oval, round, or filamentous. morbilliform rash may be seen in children receiving cefaclor. Stevens-
Bacterial resistance. The major mechanism of resistance to Johnson syndrome and toxic dermal necrolysis have been reported.
cephalosporins is the microbial elaboration of various β-lactamases Anaphylactic reactions to cephalosporins seem to be rare, with
(cephalosporinases). First-generation agents are very sensitive to an incidence ranging from 0.0001% to 0.1% of individuals exposed.
β-lactamase hydrolysis, with the second to fifth generations more In 9388 patients without a history of penicillin allergy given cepha-
resistant to β-lactamases. losporins, two anaphylactic reactions were reported (0.2%). In a ret-
Absorption, fate, and excretion. Oral cephalosporins are gen- rospective study of 350,000 adverse drug reactions, six fatal cases of
erally well absorbed, with all except cefadroxil and cefprozil having cephalosporin-induced anaphylaxis were reported, with three of the
their absorption delayed, but not reduced, by food. Cephalosporins six cases in patients with a history of penicillin allergy.
are hydrophilic and widely distributed in extracellular fluid, but they The issue of cross-sensitivity between the cephalosporins and pen-
do not enter the cells of the immune system (macrophages, polymor- icillins has never been satisfactorily resolved. Estimates range from
phonuclear leukocytes), as do lipophilic macrolides, tetracyclines, 1.1% (the same as the allergy incidence to cephalosporins in the gen-
and lincosamides. Plasma protein binding is 10% for cephalexin, eral population) to 18% in the earliest studies. Penicillin-allergic indi-
25% for cefaclor, 8% to 17% for cephradine, and 80% to 90% for viduals may have a fourfold greater risk of allergy to cephalosporins
cefazolin and cefoxitin. Excretion of most cephalosporins takes place than individuals not allergic to penicillins; however, penicillin-allergic
mainly by glomerular filtration and active tubular secretion in the individuals have a three to four times greater risk of allergy to any drug.
kidney. The serum half-life is 50 to 80 minutes for cephalexin, 48 No skin test is available to detect cephalosporin allergy, and experience
to 80 minutes for cephradine, and 35 to 54 minutes for cefaclor. In with desensitization is limited and not standardized.
patients with end-stage renal disease, these half-lives may increase to Cephalosporins are generally contraindicated in patients with a
19 to 22 hours for cephalexin, 8 to 15 hours for cephradine, and 2 to positive penicillin skin test to the minor determinant mixture or a
3 hours for cefaclor. history of local or systemic penicillin anaphylaxis (severe urticaria,
472 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

bronchospasm, hypotension, exfoliative dermatitis), unless cephalo-


TABLE 33-7  Macrolide Preparations
sporins are mandated in the management of a life-threatening infec-
tion, and anaphylaxis antidotal therapy is readily available.
­Available in the United States
Drug interactions. Antacids, H2 histamine receptor antagonists, Nonproprietary (Generic) Name Proprietary (Trade) Name
proton pump inhibitors, as well as iron supplements may reduce Erythromycin base (film-coated) Erythromycin Filmtabs
the oral absorption of some cephalosporins. Food decreases the oral Erythromycin (enteric-coated) E-Base, E-Mycin, Ery-Tab, Eryc
absorption of cefuroxime and cefpodoxime. Some cephalosporins Erythromycin stearate Erythrocin stearate
such as cefotetan may induce a disulfiram reaction with ethanol and Erythromycin ethylsuccinate E.E.S., EryPed
may cause hypoprothrombinemia. Nephrotoxicity may be seen with Erythromycin lactobionate —
the combination of cephalosporins with aminoglycosides or loop Clarithromycin Biaxin
diuretics. Azithromycin Zithromax
Contraindications. Cephalosporins are contraindicated in patients Dirithromycin Dynabac
allergic to these drugs and in individuals with a history of severe Troleandomycin TAO
penicillin reactions or a positive skin test reaction to the penicillin
minor determinant mixture.
O
Other β-lactam antibiotics
H3C CH3
Carbapenems. Carbapenems are derivatives of thienamycin (from
Streptomyces cattleya) and differ from penicillins by the replacement OH OH H3C
H3C CH3 HO
of the sulfur by a methylene group in the five-membered ring of the N CH3
OH
β-lactams (see Fig. 33-7). Currently, four carbapenems are available
H 3C
for parenteral use in the United States: imipenem, meropenem, O O
ertapenem, and doripenem. Imipenem is combined with cilastatin to O
CH3
reduce the hydrolysis of imipenem by renal dehydropeptidase. O O O CH3 CH
3
Carbapenems have a very wide antibacterial spectrum, have a high CH3
specificity for PBP2 of gram-positive and gram-negative microorgan-
OH
isms (resulting in ovoid organisms), and are not hydrolyzed by most
H 3C O CH3
β-lactamases. Their indications are listed in Table 33-6.
Microbial resistance to carbapenems is via the loss of an outer
membrane protein, resulting in retarding cell wall penetration of the Erythromycin
drugs, altered PBPs in Enterococcus faecium and MRSA, and hydrolysis FIG 33-8  Structure of erythromycin.
by metallo β-lactamases and other β-lactamases.
Carbapenems are classified as FDA pregnancy category B or C erythromycin A and a 14-membered macrolide with a 3-keto group
drugs, are cross-allergenic with other β-lactams, may increase the level substitution.
of serum liver transaminases, may induce PMC, and are associated
with a 3% to 4% incidence of skin rash. Imipenem, meropenem, and Mechanism of action and antibacterial spectrum
ertapenem are associated with increased central nervous system (CNS) The mechanism of action of macrolides is to bind reversibly to the P
toxicity and seizures. Doripenem has a lower risk of CNS toxicity. site of the 50S ribosomal subunit and inhibit RNA-dependent pro-
Monobactams. Aztreonam is a monocyclic β-lactam (monobac- tein synthesis by stimulating the dissociation of peptidyl transfer
tam) lacking the thiazolidine ring of penicillin (see Fig. 33-7). It is RNA (tRNA) from the ribosome (Fig. 33-9). Erythromycin is active
available only parenterally. It does not bind to the PBPs of gram-pos- against gram-positive aerobic/facultative staphylococci and strepto-
itive or anaerobic microorganisms; its spectrum is limited to aerobic cocci, gram-negative anaerobes (M. catarrhalis, Bordetella pertussis,
gram-negative species. Specific organisms are listed in Table 33-6. Legionella pneumophila), and Mycoplasma pneumoniae. It is also active
Aztreonam is not the initial drug of choice for any infection. It is not against organisms listed in Table 33-8 (see also Table 33-5).
indicated in dental infections. Microorganisms generally resistant to macrolides include H. influ­
Aztreonam is an FDA pregnancy category B drug and lacks cross-al- enzae, Peptostreptococcus, Aggregatibacter actinomycetemcomitans,
lergenicity with β-lactams. Aztreonam induces β-lactamase produc- Pasteurella, Fusobacterium, Mycobacterium tuberculosis, MRSA, and
tion and may be synergistic with the renal toxicity and ototoxicity of Enterobacteriaceae. Marginally affected organisms include Prevotella
aminoglycosides. and Porphyromonas.
Clarithromycin and azithromycin have special indications because
Macrolide and Ketolide Antibiotics they are more active against some organisms than is erythromycin, or
Chemistry and classification (Table 33-7; Fig. 33-8) in some cases in which erythromycin is not active. (See Table 33-5.)
Macrolide antibiotics are characterized by large 14-membered,
15-membered, or 16-membered lactone rings. Erythromycin, as Bacterial resistance
derived from Streptomyces erythreus, was introduced in 1952, and The major mechanism for microbial resistance is demethylation of the
azithromycin and clarithromycin were introduced in 1991 and 1992, 2058 residue of the gene coding for the 23S ribosomal RNA peptidyl
respectively. Azithromycin is a 15-membered macrolide with an transferase region, resulting in reduced macrolide binding (ribosomal
added nitrogen and N-methylation (making it technically an azalide), protection). The erm (erythromycin-resistant methylase) gene respon-
whereas clarithromycin is formed by the alkylation of a hydroxyl group sible for the ribosomal protection is often associated with tetracycline
of erythromycin (a 14-membered ring). Troleandomycin is a synthetic resistance genes and is frequently combined with the resistance genes
derivative of oleandomycin, dirithromycin is a prodrug yielding eryth- for lincosamides (clindamycin) and streptogramins (quinupristin-dal-
romycylamine in the intestine, and telithromycin is a derivative of fopristin) to form the macrolide-lincosamide-streptogramin B (MLSB)
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 473

Growing polypeptide

Chloramphenicol
binds to 50S portion inhibiting
formation of peptide bond

50S
portion

Tetracycline
interferes with attachment of
tRNA to mRNA-ribosome complex

Macrolides
Linezolid bind to 50S portion preventing
Binds to 50S portion translocation movement of
and inhibits initiation ribosome along mRNA
of protein synthesis

mRNA

Direction of
30S ribosome travel
portion
Aminoglycosides
bind to 30S portion inhibiting
translation and causing code
on mRNA to be read incorrectly
70S bacterial
ribosome
FIG 33-9  Sites of inhibition of bacterial ribosomal protein synthesis by various drugs.

TABLE 33-8  Macrolides as Agents of Absorption, fate, and excretion


Choice or Alternative Drugs* Erythromycin and azithromycin are available for oral and intravenous
use, whereas the remaining agents are used only orally. Bioavailability
Drugs of Choice Alternative Agents
ranges from 10% for dirithromycin to 40% to 50% for azithromycin,
Bartonella henselae Actinomyces israelii clarithromycin, and erythromycin. Food in the stomach may increase
Bartonella quintana Bacillus anthracis or have no effect on the absorption of azithromycin, but the remain-
Bordetella pertussis Borrelia burgdorferi ing macrolides should be taken 1 hour before or 2 hours after a meal,
Campylobacter jejuni Capnocytophaga canimorsus except for estolate and ethylsuccinate salts of erythromycin, which may
Chlamydia trachomatis Chlamydia pneumoniae be taken without regard to meals. (Note, however, that the estolate
Corynebacterium diphtheriae Eikenella corrodens form of the drug has a higher risk of hepatic toxicity.)
Corynebacterium jeikeium Erysipelothrix rhusiopathiae Erythromycin base is poorly resistant to gastric acid and is
Haemophilus ducreyi Leptotrichia buccalis prepared with enteric coating or as various salts (stearate), esters
Helicobacter pylori Mycobacterium leprae (ethylsuccinate), or salts of esters (estolate), which protect against
Legionella pneumophila Streptococcus pneumoniae gastric acid degradation. Macrolides are best absorbed in the small
Mycobacterium avium Streptococcus pyogenes intestine, and when given orally in standard doses, generally pro-
Mycobacterium kansasii duce adequate tissue MIC concentrations. Erythromycin may have
Mycobacterium marinum a variable absorption rate. The time to maximum blood concentra-
Mycoplasma pneumoniae tions of the macrolides after oral administration is about 2 hours.
Ureaplasma urealyticum Impaired renal function may reduce the excretion of the macrolides,
*Specific macrolide(s) for most indications are listed in Table 33-5.
with the elimination half-life of erythromycin increasing from 1.6
Drug combinations are used in some cases. to 5-6 hours in anuric patients. Clarithromycin and erythromycin
Abramowicz M, Zuccotti G, editors: Handbook of antimicrobial drugs: are primarily eliminated in the urine, and azithromycin is primar-
drugs for bacterial infections, The Medical Letter ed 20, 2015, New ily eliminated in the bile, although erythromycin may also undergo
Rochelle, NY. significant biliary excretion. The average serum half-lives are
∼68 hours for azithromycin, 3 to 7 hours for clarithromycin, and
aggregate, which confers resistance to all three antibiotic groups simul- ∼1.6 hours for erythromycin.
taneously. Other macrolide resistance mechanisms include active A remarkable property of macrolides and highly fat-soluble tetracy-
efflux genes encoding for transport efflux proteins and an esterification clines and clindamycin is selective uptake by phagocytic cells and fibro-
gene that codes for inactivation of the macrolides by phosphorylation blasts, which function as repository drug depots and as a drug delivery
and glycosylation. system to areas of inflammation and infection. These cells concentrate
474 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

the macrolides and then transport them to areas of tissue pathology receptor. Motilin is a regulatory peptide of the gastrointestinal tract
where they are released for their antiinfective and antiinflammatory that stimulates enzyme secretions by the stomach and pancreas and
properties. The tissue concentrations of azithromycin may reach 100 induces strong phasic contractions of the stomach. The most sig-
to 1000 times that of blood and persist long after blood levels have nificant agonists of the motilin receptor are the 14-membered mac-
declined because of their significant post-antibiotic effects. The tissue rolides (erythromycin); azithromycin and clarithromycin are lesser
concentration of azithromycin may exceed the microorganism’s MIC stimulants.
for 2 to 10 days, and the elimination half-life in abscesses may be 4 days. Approximately 30 cases have been reported of macrolide-induced
hearing loss, with at least two cases judged to be irreversible. Most
General therapeutic uses seem to be associated with erythromycin doses exceeding 4 g/day or
Macrolides are often indicated for treating community-acquired bac- from accumulation of lesser doses in patients with impaired renal or
terial pneumonia because of their action against numerous causative hepatic function. In patients with renal impairment, the dose of eryth-
organisms. Microbial resistance is becoming increasingly common, romycin should be no greater than 1.5 g/day. Ototoxicity is particularly
however. They are useful against various chlamydial infections and common in patients with HIV/AIDS because of the use of macrolides
numerous gram-positive coccal infections, and they are also active as prophylaxis for Mycobacterium avium infections. All macrolides
against Corynebacterium (see Table 33-8). The 14-membered macro- induce ototoxicity, possibly by an effect on the auditory nerve path-
lides possess antiinflammatory effects distinct from their antimicrobial ways, and tinnitus has been observed even with therapeutic doses of
actions by decreasing proinflammatory cytokine release from phago- azithromycin.
cytes, which may prove useful in management of rheumatoid arthritis, Long QT syndrome is characterized by delayed ventricular repo-
cystic fibrosis, asthma, and chronic sinusitis. larization that triggers ventricular tachyarrhythmias, most notably
torsades de pointes, resulting in syncope, seizures, or sudden death.
Therapeutic uses in dentistry Long QT syndrome may be either congenital or acquired, and the
Erythromycin has a long and successful history of use against acute list of drugs, diseases, and metabolic disorders inducing torsades de
orofacial infections, particularly in β-lactam–allergic patients. Its pointes is long and impressive. Susceptible patients are at greater risk
spectrum of activity is good to excellent against gram-positive aero- of drug-induced torsades.
bic/facultative cocci (streptococci, some staphylococci). Its spectrum At least four cases of acute pancreatitis associated with erythromy-
is not generally favorable against gram-negative anaerobes associ- cin have been reported, and several cases of mania have been asso-
ated with orofacial infections, including Prevotella, Porphyromonas, ciated with clarithromycin in patients with and without HIV/AIDS.
Fusobacterium, and Veillonella. Azithromycin has been observed to Stevens-Johnson syndrome (erythema multiforme) may occur with
be effective against oral spirochetes and pigmented anaerobes. In the erythromycin along with a hypersensitivity syndrome associated with
management of acute periapical abscesses, azithromycin, 500 mg/day azithromycin and clarithromycin consisting of fever, rash, hepatitis,
for 3 days, has shown comparable efficacy to amoxicillin/clavulanic interstitial nephritis, oliguria, and xerostomia. Azithromycin and
acid, 625 mg three times daily for 5 to 10 days. Macrolides are also use- erythromycin are classified as FDA pregnancy category B drugs, and
ful for endocarditis prophylaxis. Due to its associated hepatic toxicity, clarithromycin is classified as an FDA pregnancy category C drug.
there is no reason for using the estolate form of erythromycin in dental
applications. Drug interactions
Clarithromycin is most active against gram-positive anaerobes Erythromycin and clarithromycin, primarily through their inhibition
(Actinomyces, Propionibacterium, Lactobacillus), whereas erythromycin of the liver microsomal enzyme drug metabolizing and secondarily
is more active than azithromycin for these organisms. Azithromycin through their effect on gastrointestinal microbial flora, increase serum
has the best activity against gram-negative anaerobes (Fusobacterium, levels of fluconazole, ranitidine, alfentanil, benzodiazepines, tacrolimus,
Prevotella, Porphyromonas, Wolinella, Selenomonas, and A. actinomyce­ theophylline, vinblastine, bromocriptine, buspirone, carbamazepine,
temcomitans). Azithromycin may be more active against streptococci cyclosporine, digoxin, disopyramide, ergot alkaloids, felodipine, oral
and staphylococci than erythromycin and clarithromycin and has anticoagulants, methylprednisolone, and omeprazole. Azithromycin has
much less propensity for drug interactions. Prolonged use of erythro- much less effect on the liver microsomes. Macrolide blood levels may
mycin and possibly other macrolides may lead to superinfection with be increased by fluconazole and decreased by theophylline. Antacids
gram-negative enteric bacilli. reduce the rate, but not the total amount, of macrolide absorption, and
the combination of macrolides and oral contraceptives may result in
Adverse effects cholestasis. Bacteriostatic macrolides may interfere with the bactericidal
Serious toxicity with macrolides is rare but occasionally significant. effect of cell wall inhibitors. Concomitant administration with a fluoro-
The most important adverse effects include epigastric pain, ototoxicity quinolone, or pimozide, may lead to torsades de pointes.
(deafness), ventricular arrhythmias (torsades de pointes), acute pan- After only 3 days of administration, macrolides may seriously
creatitis, mania, cholestatic hepatitis, hypersensitivity syndrome, and reduce digoxin metabolism in the gastrointestinal tract by Eubacterium
certain drug interactions. lentum, resulting in digitalis toxicity because the microorganism may
Cholestatic hepatitis is much more common with the estolate metabolize 30% to 40% of the drug. Macrolides may potentiate the
form of erythromycin than with other forms of erythromycin or anticoagulant effect of oral anticoagulants. Concomitant use of mac-
other macrolides. This reaction can be misdiagnosed as viral hepatitis. rolides may increase the myopathy and rhabdomyolysis seen with the
Symptoms usually appear after approximately 10 days of erythromycin “statin” anti-cholesterol agents.
use, disappear 2 to 4 weeks after drug discontinuance with no residual
effects, and readily reappear with drug readministration. This reaction Contraindications
is less common in children. Macrolides are contraindicated in patients with allergy to the drugs
The most common serious adverse reaction associated with mac- in patients with a history of previous allergic cholestatic hepatitis.
rolides, particularly erythromycin, is potentially severe epigastric Macrolides are also contraindicated in combinations with other drugs
pain resulting from stimulation of the gastric smooth muscle motilin with which they interact, such as those that induce torsades de pointes.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 475

The maximum daily dose should be 4 g in adults with normal renal some staphylococcal strains by a nucleotidyl transferase (drug inacti-
function and 1.5 g/day in patients with impaired renal function. vation). Adenine methylation is plasmid mediated and confers MLSB
resistance. The M phenotype macrolide resistance in S. pneumoniae
Ketolides does not confer resistance to clindamycin. If erythromycin resistance
Ketolides (telithromycin) are derivatives of erythromycin A specifically in staphylococci is inducible and not constitutive, the microorganisms
designed for activity against bacteria responsible for community-acquired are resistant only to the 14-membered and 15-membered macrolides
respiratory tract infections. Telithromycin is a 14-membered macrolide and remain sensitive to lincosamides, streptogramins, and 16-mem-
with a 3-keto group substitution. The oral bioavailability of telithromy- bered macrolides. Constitutive resistance in staphylococci of the MLSB
cin is approximately 55%, with maximum serum concentrations at 1 to type confers resistance to all these antibiotics simultaneously.
3 hours. The elimination half-life is about 13 hours; the drug has a long
post-antibiotic effect and is highly concentrated in white blood cells and Absorption, fate, and excretion
pulmonary tissue. It is primarily metabolized in the liver. Telithromycin Clindamycin is well absorbed orally with a 90% bioavailability not
inhibits bacterial protein synthesis by binding to the 50S ribosomal sub- appreciably reduced by food. The time to oral peak serum levels is 45
unit to inhibit translation at the peptidyl transferase site. The drug also to 60 minutes, with an elimination half-life of 2.4 to 3 hours. With renal
inhibits formation of the bacterial 50 and 30S ribosomal subunits. failure the elimination half-life increases to 6 hours, with a doubling of
Telithromycin is active against a wide spectrum of respiratory the serum level. The drug penetrates well into bone but not cerebro-
pathogens, including S. pneumoniae, H. influenzae, M. catarrhalis, C. spinal fluid; is metabolized primarily in the liver (>90%); and is highly
pneumoniae, M. pneumoniae, L. pneumophila, group A and B strepto- concentrated in the bile, where it may alter colonic flora for 2 weeks
cocci (S. pyogenes and Streptococcus agalactiae). after it is discontinued. In a manner similar to macrolides, clindamy-
The most frequent adverse reactions associated with telithromycin cin is concentrated preferentially in polymorphonuclear cells, alveolar
are diarrhea (12% to 20%), nausea (2% to 12%), dizziness (2% to 5%), macrophages, and abscess tissue.
and headache (2.5% to 5%). A major concern is the drug’s association
with liver toxicity. Telithromycin is an inhibitor of the liver microsomal General therapeutic uses
enzyme cytochrome P450 system and would be expected to increase blood Clindamycin is used in the treatment of certain infections caused by
levels of many drugs. Similar to macrolides, telithromycin may prolong susceptible strains of streptococci, staphylococci, pneumococci, or
the QT interval. It is classified as an FDA pregnancy category B drug. anaerobes such as Bacteroides. Clindamycin may be indicated in the
Telithromycin has no place in the management of acute or chronic treatment of refractory bone infections. Clindamycin is also useful in
orofacial infections unless dictated by sensitivity testing. Its use is lim- treating certain conditions involving anaerobes, such as infections of
ited in the United States to community-acquired bacterial pneumonia. the female genital tract, pelvic infections, and abdominal penetrat-
ing wounds (see Table 33-5). It can also be used in combination for
Lincosamides Pneumocystis carinii and for toxoplasmosis.
Clindamycin and lincomycin are the only lincosamide antibiotics.
Lincomycin was isolated from Streptomyces lincolnensis in 1962, and Therapeutic uses in dentistry
clindamycin (7-chloro-7-deoxy-lincomycin) was introduced in 1966. Although amoxicillin and penicillin V remain drugs of choice for acute
Clindamycin (Fig. 33-10) is used almost exclusively because of its orofacial infections, a resurgence in clindamycin use may be appropri-
greater efficacy and superior pharmacokinetics. ate as the oral microbial resistance to β-lactams continues to increase.
It is anticipated that oral microbial resistance to clindamycin will also
Mechanism of action and antibacterial spectrum increase proportionately along with the specter of MLSB resistance
The receptor site for lincosamides is identical to that of macrolides, shared with macrolides and streptogramins.
chloramphenicol, and streptogramins: the 23S subunit of the 50S bac-
terial ribosome, resulting in bacteriostatic inhibition of microbial Adverse effects
protein synthesis. Clindamycin has significant activity against many Minor adverse reactions associated with clindamycin include nau-
gram-positive and gram-negative anaerobic and facultative/aero- sea and vomiting, abdominal pain, esophagitis, glossitis, stomatitis,
bic microorganisms. The major indications are listed in Table 33-9.
Clindamycin has indications for some oral infections.
TABLE 33-9  Indications for Clindamycin*
Bacterial resistance
Resistance to clindamycin occurs by three mechanisms: (1) alteration Methicillin-sensitive Staphylococcus aureus
of 23S ribosomal RNA of the 50S ribosomal subunit by adenine meth- Streptococcus pyogenes
ylation (ribosomal protection), (2) an altered single 50S ribosomal Streptococcus pneumoniae
protein at the receptor site (receptor alteration), or (3) inactivation in VGS
Peptostreptococcus
Cl
Bacillus anthracis
CH3 O H CH3 Clostridium perfringens
N Actinomyces israelii
N
O CH3 Leptotrichia buccalis
H Fusobacterium species
HO S
Capnocytophaga canimorsus
H3C Bacteroides (oropharyngeal strains)
HO OH
*Abramowicz M, Zuccotti G, editors: Handbook of antimicrobial drugs:
Clindamycin drugs for bacterial infections, The Medical Letter ed 20, 2015, New
FIG 33-10  Structure of clindamycin. Rochelle, NY.
476 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

allergy, reversible increase in serum transaminase levels, reversible involved in acute orofacial infections, periodontitis, and acute nec-
myelosuppression, metallic taste, maculopapular rash (3% to 10%), rotizing ulcerative gingivitis.
and diarrhea (2% to 20%, average of 8%). High intravenous doses of
clindamycin may result in a neuromuscular blockade similar to that of Mechanism of action and antibacterial spectrum
aminoglycosides. The antimicrobial activity of metronidazole requires entry into the cell
The major concern with clindamycin has been its purported pro- and reduction of its nitro group to produce metabolites that damage
pensity to cause antibiotic-induced diarrhea and colitis, most nota- DNA, eventually inducing cell death. Metronidazole is active only
bly PMC, based on early reports of incidences reaching 10%. It is now against bacteria that are obligate anaerobes. It is a concentration-
apparent that the association of clindamycin with these colonic disor- dependent rather than time-dependent antibiotic. Because metroni-
ders in outpatient use is less than previously reported, although none- dazole metabolites interfere with nucleic acid synthesis, concerns have
theless real. Although antibiotic-associated diarrhea is common in the been raised regarding its potential for mutagenicity, carcinogenicity,
outpatient environment and readily managed by drug discontinuance, and teratogenicity.
serious antibiotic-induced colitis and potentially lethal PMC is more Metronidazole penetrates all bacterial cells equally well. In sensitive
rare. anaerobes, the nitro moiety of the drug is enzymatically reduced, how-
Care should be taken with patients who have recently recovered ever, and this metabolite is the active form of the drug. Metronidazole
from C. difficile–associated diarrhea or colitis for 2 months after is almost always bactericidal. The drug reacts with bacterial DNA,
the cessation of the disease. Any elective dental procedure requiring causing inhibition of DNA replication, fragmentation of existing DNA,
antibiotic therapy or prophylaxis is best postponed for this 2-month and in low doses, mutation of the bacterial genome.
period. If antibiotic therapy is required, antibiotics far less associated
with antibiotic-induced diarrhea (e.g., penicillin V, macrolides) are Bacterial resistance
appropriate. Microbial resistance to metronidazole is limited probably because
of its limited clinical use, except in developing countries, where it is
Drug interactions widely used to manage parasitic diseases. A notable exception to this
Clindamycin acts synergistically with nondepolarizing (curare-like) generalization is the high level of resistance in H. pylori in developed
neuromuscular blocking drugs in blocking neurotransmission at skel- countries. Resistance to metronidazole is chromosomally mediated
etal muscle. Oral absorption of clindamycin is slowed by kaolin-pectin and plasmid-mediated by a reduction in activity or expression of sev-
antidiarrheal drugs. eral genes (rdxA, nimA, nimB) that control nitroreductase activity,
which reduces the concentration of active metronidazole metabolites
Contraindications within the microbial cell.
Clindamycin is contraindicated in patients allergic to the drug and in Subinhibitory concentrations of metronidazole may increase resis-
combination with curare-like neuromuscular blocking drugs. All anti- tance rates in various periodontal pathogens, including Fusobacterium,
biotics should be avoided if possible for 2 months after antibiotic-in- Prevotella, Porphyromonas, and Peptostreptococcus. Bacteroides strains
duced colitis. resistant to metronidazole also acquire enhanced virulence properties.

Metronidazole Absorption, fate, and excretion


Metronidazole (Fig. 33-11) is a synthetic nitroimidazole patterned Metronidazole is almost completely absorbed from the gastrointestinal
after a naturally occurring antiparasitic substance that was isolated tract (oral bioavailability approaches 100%) so that serum levels are
from a Streptomyces species in 1955. The drug was introduced into essentially the same whether the drug is administered orally or intra-
medicine in 1959 and was quickly found to possess strong tricho- venously. Food may delay peak serum levels of metronidazole but not
monacidal activity. Since then, metronidazole has become the drug the total amount absorbed. Metronidazole attains a peak blood level
of choice for various protozoal infections. A chance observation orally in 1 to 2 hours, has a wide volume of distribution, has excellent
that the symptoms of acute necrotizing ulcerative gingivitis were CNS penetration, has an elimination half-life of 8 hours, and is bio-
relieved in a woman receiving metronidazole for the treatment of transformed into five metabolic products, all of which have anti-an-
vaginal trichomoniasis stimulated research on the drug’s antibac- aerobic activity. The pharmacokinetics of metronidazole are the same
terial effects, culminating in its approval in 1981 for the treatment in pregnant and nonpregnant women, its metabolism is reduced in the
of anaerobic bacterial infections. Its extensive use in treating para- presence of severe hepatic dysfunction, and its pharmacokinetics are
sitic diseases worldwide has led to significant resistance to the drug not significantly altered with renal impairment.
where parasites are a major problem. It was soon discovered that
the drug possessed exceptional activity against obligate anaerobic General therapeutic uses
and microaerophilic microorganisms, including microorganisms The principal medical indications for metronidazole are anaerobic
abdominal and CNS infections, bacterial vaginosis, protozoan and H.
pylori infections, and the management of C. difficile–associated diar-
OH
rhea and colitis. Metronidazole is very active against obligate anaer-
obes (e.g., Bacteroides, Porphyromonas, Prevotella, Fusobacterium,
Peptostreptococcus, Clostridium), many of which are associated with
O
periodontitis, and various human parasites (e.g., Trichomonas vaginalis,
N
N+ CH3 Gardnerella vaginalis, Entamoeba histolytica, Balantidium coli). A. acti­
–O nomycetemcomitans, E. corrodens, Actinomyces, and Propionibacterium
N are commonly resistant to metronidazole. The combination of metro-
nidazole with amoxicillin may significantly enhance its activity against
Metronidazole A. actinomycetemcomitans, apparently by increasing cellular uptake of
FIG 33-11  Structure of metronidazole. metronidazole.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 477

Therapeutic uses in dentistry H. pylori to metronidazole and macrolides, tetracyclines have gained
Metronidazole is highly effective against gram-negative anaerobic importance in the treatment and prevention of peptic ulcers and gas-
pathogens responsible for acute orofacial infections and chronic peri- tric cancer and have emerged as a prophylactic agent in the prevention
odontitis. Combination of metronidazole with a β-lactam antibiotic of multidrug-resistant malaria in travelers and the management of
for oral infections may be indicated for serious acute orofacial infec- community-acquired pneumonia, particularly in penicillin-resistant
tions and in the management of aggressive periodontitis. and macrolide-resistant strains.
Metronidazole is a concentration-dependent, not time-dependent, Tetracyclines comprise a group of antibiotics with a similar anti-
antibiotic. The promiscuous use of metronidazole for classic chronic bacterial spectrum but differing pharmacokinetic properties created by
periodontitis is a misuse of the drug and may contribute to the increas- various chemical substitutions on the hydro-naphthacene four-ringed
ing resistance of metronidazole seen with parasites, H. pylori, and other nucleus. Chlortetracycline, oxytetracycline, tetracycline, and demeclo-
microorganisms. cycline constitute the first-generation tetracyclines. Second-generation
agents introduced from 1965 to 1972 include minocycline, methacy-
Adverse effects cline, and doxycycline. Glycylcyclines are third-generation agents. The
Minor adverse reactions associated with metronidazole include revers- first microorganism clinically detected to be resistant to tetracyclines
ible neutropenia, metallic taste, dark or red-brown urine, skin rash, was a Shigella dysenteriae strain in 1953. Today, doxycycline is the
urethral or vaginal burning sensation, gynecomastia, and nausea and most commonly used tetracycline (Table 33-10).
vomiting. Rare major adverse reactions include pancreatitis; PMC;
peripheral neuropathy; disulfiram reaction when combined with Mechanism of action
ethanol; and CNS toxicity consisting of seizures, encephalopathy, cer- Tetracyclines inhibit bacterial protein synthesis by preventing the
ebellar dysfunction, paresthesias, mental confusion, and depression. association of aminoacyl-tRNA with the bacterial ribosome. The
These neurologic reactions generally occur only with high, prolonged, drugs must transverse the gram-negative microbial outer membrane
cumulative doses. via porin channels or through the gram-positive cell wall in its electro-
Because metronidazole affects DNA synthesis, numerous studies negative hydrophobic form and attach to a single high-affinity binding
have addressed its potential to cause birth defects. Its use in pregnancy site on the ribosomal 30S subunit and protein 7 on the 16S rRNA base.
does not seem to be associated with any congenital abnormalities,
preterm delivery, or low birth weight in newborns, and the drug has an Bacterial resistance
FDA pregnancy category B classification. Microbial resistance to tetracyclines is widespread, transposable,
inducible, and commonly permanent because their resistance genes
Drug interactions are almost always combined in transposable elements with the genes
Barbiturates may reduce the efficacy of metronidazole, and cimeti- for resistance to other antibiotics (multidrug resistance gene cassettes).
dine may reduce its liver metabolism. The concurrent use of metroni- Of the three mechanisms for tetracycline resistance (drug efflux, ribo-
dazole and ethanol may result in acute psychosis and the disulfiram somal protection, and enzymatic inactivation), drug efflux is the most
reaction (flushing, tachycardia, nausea and vomiting), although for important, with at least 300 different active efflux proteins capable of
most individuals the risk is minor. Metronidazole may increase lith- extruding tetracycline from the bacterial cell.
ium blood levels, decrease the body clearance of phenytoin, and signifi- Tetracyclines are one of the most active chemical inducers of micro-
cantly increase blood warfarin levels by decreasing its liver metabolism. bial resistance gene expression and downregulate a repressor gene that
controls efflux activity for not only tetracyclines but also possibly other
Tetracyclines and Glycylcyclines antibiotics. Only nanomolar amounts of tetracycline are necessary to
Tetracyclines (Fig. 33-12) are a group of broad-spectrum, bacterio- de-repress this system and greatly increase antibiotic efflux from bac-
static antibiotics that have been extensively used in the treatment of terial cells. Tetracyclines also promote the mobility of resistance deter-
numerous and varied infections. Their widespread use, and often minants (transfer of resistance genes between bacteria) by stimulating
misuse, has resulted in the appearance of many bacterial strains that the frequency of bacterial conjugation. Considering these extraordi-
are resistant to these drugs, which has curtailed their clinical useful- nary properties of tetracyclines to induce and promote microbial resis-
ness. Paradoxically, the clinical use of tetracyclines has had a recent tance not only to themselves but also other antibiotics, it would seem
resurgence of interest with the growing realization that the drugs may prudent to restrict their use to serious medical infections and restrict
be lifesaving in the treatment of serious nosocomial infections from their use in most cases of periodontitis, where they may have limited or
highly and multiply antibiotic-resistant methicillin-resistant staphy- even undocumented value.
lococci. This renewed effectiveness of tetracyclines may be related to
the almost complete lack of use of the drugs in hospitals for several
decades, possibly leading to the loss of tetracycline resistance genes in TABLE 33-10  Commercially Available
this environment. Because of the advent of widespread resistance of ­Tetracycline and Glycylcycline Preparations
Nonproprietary (Generic) Name Proprietary (Trade) Name
OH O OH O O Tetracycline hydrochloride Tetralen, Panmycin, Sumycin,
OH
Tetracyn, Tetracap
2 NH2 Oxytetracycline hydrochloride Terramycin
7 6 5 Demeclocycline hydrochloride Declomycin
OH
H H Doxycycline hyclate Vibramycin, Doxy Caps
CH3 OH N Minocycline hydrochloride Minocin, Vectrin, Dynacin
H3C CH3
Glycylcycline
Doxycycline
Tigecycline Tygacil
FIG 33-12  Structure of doxycycline.
478 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

Absorption, fate, and excretion major drugs in treating rickettsial diseases, Mycoplasma, Chlamydia
Tetracyclines are adequately but variably absorbed from the gastroin- (treatment of community-acquired pneumonia), H. pylori, and Borrelia.
testinal tract with significant differences in bioavailability, as follows: (Borrelia burgdorferi is the causative agent of Lyme disease.)
chlortetracycline, 30%; demeclocycline, tetracycline, and oxytetracy- Many other bacteria nominally within their range of activity should
cline, 60% to 80%; and minocycline or doxycycline, 95% to 100%. not be treated, at least initially, with a tetracycline because of the
Dairy products, Ca2+, Mg++ and aluminum compounds, and Na+ bicar- numerous resistant strains.
bonate significantly impair tetracycline absorption either by chelation Tetracyclines have been used in the treatment of acne using oral and
or by altered gastric pH. Their serum protein binding ranges from 20% topical preparations. Tetracyclines are concentrated in the skin and are
to 40% for oxytetracycline to 80% to 95% for doxycycline, and the per- effective against Propionibacterium acnes and may have an antisebor-
cent excreted unchanged in the urine ranges from 70% for oxytetracy- rheic action to reduce skin lipids. Because tetracyclines are deposited
cline to 30% to 42% for doxycycline and 12% to 16% for minocycline. in bone, they can be used to measure the rate of bone growth. Other
With renal impairment, only doxycycline and minocycline do not have indications include plague, tularemia, cholera, brucellosis, and certain
increased half-lives and can be administered with reasonable safety. protozoal infections.
Other tetracyclines may accumulate in conditions of renal impairment,
resulting in high blood levels and possible liver necrosis and death. Therapeutic uses in dentistry
Tetracyclines are metabolized in the liver to a varying degree The use of tetracyclines in the management of acute orofacial infec-
depending on the individual drug and are highly concentrated in the tions is widely considered inappropriate because of their bacteriostatic
bile at levels three to five times higher than serum. More recent tetracy- activity and extensive microbial resistance. Evidence does not support
clines are more lipid-soluble with greater tissue distribution than ear- the use of tetracyclines in the management of chronic periodontitis.
lier tetracyclines. Enterohepatic circulation and incomplete absorption Data showing clinical efficacy are lacking, and there is a propensity to
may lead to high drug concentrations in the feces, particularly with induce microbial resistance gene expression and stimulation of drug
older agents. Doxycycline may be found to some degree in the feces efflux mechanisms and common association with multiple resistance
as an inactive form, and it is as yet unknown if this metabolic product genes to other antibiotics in transposable elements. Tetracyclines may
is as capable of inducing resistance gene expression or transfer as the be of use in the management of localized aggressive periodontitis and
parent compound. The serum half-lives of the various agents are 12 its associated organism, Aggregatibacter (formerly Actinobacillus) acti­
to 16 hours for oxytetracycline, tetracycline, and demeclocycline; 14 to nomycetemcomitans. Tetracyclines also seem to inhibit inflammatory
16 hours for methacycline; 11 to 18 hours for minocycline; and 15 to matrix metalloproteinase activity. Tetracyclines may also be used sub-
25 hours for doxycycline. Peak serum concentrations are reached in gingivally. The degree of benefit of the use of tetracyclines in periodon-
2 hours after the usual therapeutic doses. tal disease has to be weighed against the risk of developing resistant
strains of microorganisms.
General therapeutic uses (Table 33-11 and also see Table 33-5)
Tetracyclines are broad-spectrum antibiotics. Table 33-11 lists some sen- Adverse effects
sitive organisms against which they are used in therapy. Tetracyclines are Numerous adverse drug reactions are associated with tetracyclines.
Tetracyclines may induce photosensitivity; nephrogenic diabetes
insipidus (demeclocycline); blood dyscrasias; liver dysfunction (high
TABLE 33-11  Indications for Tetracyclines doses and especially during pregnancy); pseudotumor cerebri and
bulging fontanelles (adults and infants); C. albicans overgrowth; gas-
and Tigecycline
trointestinal difficulties (nausea, vomiting, diarrhea, pancreatitis); and
Brucella various allergic manifestations, including urticaria, serum sickness,
Borrelia burgdorferi* angioneurotic edema, and anaphylaxis.
Borrelia recurrentis Minocycline in conventional doses is associated with skin, nail,
Chlamydia pneumoniae and hair pigmentation and a systemic lupus erythematosus–like syn-
Chlamydia psittaci drome, mainly in adolescents taking the drug for acne. This syndrome
Chlamydia trachomatis is usually reversible but may require corticosteroid therapy; however,
Ehrlichia chaffeensis* the absolute risk seems to be low (52.8 per 100,000 uses). Sixteen cases
Ehrlichia ewingii* of autoimmune hepatitis have been described.
Mycobacterium marinum† Minocycline is the only tetracycline that induces vestibular tox-
Mycoplasma pneumoniae icity (ataxia, loss of balance), possibly because of its high concentra-
Rickettsiae* tion in the lipid-rich cells of the inner ear. Tetracyclines in general are
Vibrio cholerae one of the few groups of drugs that are toxic if ingested beyond their
Vibrio vulnificus expiration date, inducing a Fanconi-like syndrome (azotemia, kidney
Yersinia pestis damage). Tetracycline-induced acute interstitial nephritis may result
Indications for Tigecycline in acute renal failure. Hepatotoxicity is rare except at high doses and is
Community-acquired pneumonia caused by Streptoccocus pneumoniae, most likely to occur during pregnancy, leading to an absolute contra-
Hemophilus influenza, and Legionella pneumophila indication to the drugs in pregnancy.
Intraabdominal and skin infections due to gram-negative rods, streptococcal The chelation properties of tetracyclines are responsible for their
and staphylococcal bacteria, Kebsiella, Bacterioides, and others deposition in calcifying teeth, bone, and cartilage, and these drugs have
*Specifically doxycycline. been used as vital stains to determine bone growth. Tetracycline stain-
†Specifically minocycline. ing is not permanent in tissues that are remodeled (bone, cartilage), but
From Abramowicz M, Zuccotti G, editors: Handbook of antimicrobial it is permanent in tissues that are not remodeled (teeth). Tetracyclines
drugs: drugs for bacterial infections, The Medical Letter ed 20, 2015, should not be used in children younger than 8 years unless other anti-
New Rochelle, NY. biotics are unlikely to be effective or are contraindicated. Tetracyclines
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 479

most likely to stain the dentition severely are tetracycline and deme- marketed in 1986. Newer fluoroquinolones have improved activity
clocycline with oxytetracycline, chlortetracycline, and doxycycline (the against S. pneumoniae, S. aureus, gram-positive cocci, anaerobes, P.
least likely), but the magnitude of the staining may depend more on aeruginosa, and various other organisms.
dose and duration than the drug itself. Deposition of tetracyclines in
bone and teeth eliminates their antimicrobial activity. Because of the Mechanism of action and antibacterial spectrum (Table 33-12)
deleterious staining effects on the teeth and the liver, tetracyclines are DNA gyrase and topoisomerases are enzymes involved in the cru-
classified by the FDA as pregnancy category D drugs. Tetracyclines cial processes of DNA replication, transcription, and recombination.
should not be used in pregnancy because of staining of teeth and DNA gyrase has two subunits (A and B) regulated by two genes (gyrA
potential hepatotoxicity. and gyrB), with topoisomerase IV encoded by parC and parE genes.
Minocycline is also able to impart a grayish discoloration of Both enzymes are responsible for supercoiling DNA, forming dou-
teeth, even after tooth formation and eruption. This could at least in ble-stranded DNA, and maintaining DNA in its physiologically stable
part be an extrinsic stain; however, the exact mechanism(s) has/have and biologically active state. Topoisomerase IV nicks double-stranded
not been established. This type of staining does not occur with other DNA and seals the nicked DNA, whereas DNA gyrase guides the pas-
tetracyclines. sage of the DNA through the interior of the enzyme complex. Both
enzymes are responsible for supercoiling DNA, allowing for its fit into
Drug interactions the bacterial cell. Fluoroquinolones stabilize the enzyme complex after
Tetracyclines and all other antimicrobial ribosomal protein synthesis strand breakage and before resealing, preventing DNA supercoiling.
inhibitors may reduce the efficacy of antibiotic cell wall inhibitors, Table 33-12 indicates their antibacterial spectra and indications,
which rely on cell wall division for their action. Polyvalent cations listing organisms that are sensitive to some or all fluoroquinolones.
(aluminum, Ca2+, zinc, iron, magnesium, bismuth) may decrease gas-
tric tetracycline absorption by chelation. Na+ bicarbonate alters the Bacterial resistance
gastric pH and reduces absorption of tetracyclines. Three mechanisms account for microbial resistance to fluoroquino-
Serum levels of tetracyclines may be reduced from increased lones: mutations in DNA gyrase and topoisomerase IV, drug efflux
hepatic metabolism induced by barbiturates, carbamazepine, and pumps, and reduction in microbial outer membrane permeability. The
hydantoins. The addition of tetracyclines to coumarin anticoagulants target alterations are accomplished by simple point mutations in DNA
(e.g., warfarin) may greatly increase the latter’s effect on the INR and gyrase (gyrA) and topoisomerase IV (parC) and by efflux mechanisms.
lead to serious bleeding episodes. The effect is partially due to the
inhibitory effect of tetracyclines on the intestinal flora that produce
vitamin K. TABLE 33-12  Classification of Fluoro-
quinolones Based on Antibacterial Spectrum
Contraindications
Indications Based on Advantage of
Tetracyclines are contraindicated in children younger than 8 years, in
Group Individual Drug
cases of allergy, during pregnancy, and during lactation.
Group I (Urinary Tract Infections)
Glycylcyclines Norfloxacin (Noroxin) Gram negatives, e.g., K. pneumoniae, E. coli, P.
Glycylcyclines are synthetic derivatives of minocycline. Tigecycline is mirabilis, P. vulgaris, Providencia
the glycylcycline used in the United States. It is effective against some Group II (Broad Spectrum)
tetracycline-resistant bacteria and other organisms listed in Table Ciprofloxacin (Cipro) Pseudomonas aeruginosa, Mycobacterium tuber-
33-11. The resistance gene for glycylcyclines is carried on the same culosis, Salmonella typhi, infectious diarrhea,
transposon as the resistance genes for the macrolides and tetracycline, inhalation anthrax
making the use of glycylcyclines potentially limited. Levofloxacin (Levaquin) Pneumonia, bronchitis, urinary tract infections,
inhalation anthrax, M. tuberculosis
Fluoroquinolones Lomefloxacin (Maxaquin) Haemophilus influenzae or Moraxella catarrh-
Fluoroquinolones were introduced in the 1980s and are C6 fluorine alis respiratory tract infection, urinary tract
derivatives of nalidixic acid. The terms fluoroquinolone and quinolone infections
are often used interchangeably; however, this is technically incorrect Ofloxacin (Floxin) Pneumonia, urinary tract infections,
because nalidixic acid is not a fluoroquinolone, being devoid of the M. tuberculosis
fluorine substitution. Group III (Greater Gram-Positive Activity)
The first fluoroquinolone (norfloxacin) was synthesized in 1978
Gemifloxacin (Factive) Streptococci and other organisms causing
as a 6-fluorinated derivative of nalidixic acid with a piperazine ring
­pneumonia and bronchitis
at position 7 (Fig. 33-13). Ciprofloxacin was synthesized in 1981 and
Sparfloxacin (Zagam) Streptococci and other organisms causing
­pneumonia and bronchitis
Group IV (Greater Gram-Positive/Anti-Anaerobic Activity)
HN
Moxifloxacin (Avelox) Anaerobes including Bacteroides, ­intraabdominal
N N infections, pneumonia, skin infections, M.
tuberculosis
OH
F Organisms that are sensitive to fluoroquinolones in general include
O O
Mycoplasma pneumoniae, Legionella pneumophila, Chlamydia spe-
cies, Escherichia coli, Haemophilus influenza, Klebsiella pneumoniae,
Ciprofloxacin Neisseria gonorrhoeae, Shigella, Proteus mirabilis, and Staphylococcus
FIG 33-13  Structure of ciprofloxacin. aureus. Added indications for specific drugs are listed.
480 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

Absorption, fate, and excretion Drug interactions


Fluoroquinolones are well absorbed orally with a 70% and 90% bio- An important drug interaction with fluoroquinolones is the potential
availability for ciprofloxacin and levofloxacin. Food generally delays increase in CNS toxicity with the concomitant use of NSAIDs and
the peak concentration of ciprofloxacin and levofloxacin, but it has methylxanthines. The combination of a fluoroquinolone with tricyclic
no effect on moxifloxacin. The percent excreted by the kidneys ranges antidepressants, erythromycin, phenothiazines, and antiarrhythmic
from 27% to 73%, and protein binding ranges from 15% to 35%. agents (quinidine, procainamide, disopyramide) may increase the risk
Fluoroquinolone half-lives vary, approximately 4 hours for ciproflox- of torsades de pointes. Fluoroquinolones may reduce the liver clear-
acin and norfloxacin, 7 to 8 hours for lomefloxacin and sparfloxacin, ance of warfarin and procainamide and increase the toxicity of cyclo-
and ∼14 hours for moxifloxacin. Pharmacokinetic improvements have sporine. Omeprazole may increase fluoroquinolone blood levels, and
included longer half-lives to allow for once-daily dosing and greater antacids, sucralfate, and calcium, iron, or zinc supplements may reduce
volumes of distribution for better tissue penetration. The post-antibi- gastric absorption of fluoroquinolones.
otic effects of fluoroquinolones vary from 1 to 4 hours.
Contraindications
General therapeutic uses Ciprofloxacin should be used with caution during pregnancy and in
Table 33-12 identifies differences in indications based on groups of children. For children younger than 18 years, other fluoroquinolones
fluoroquinolones. Fluoroquinolones are used to treat urinary tract are contraindicated. Phototoxicity may occur on skin areas exposed to
infections and bacterial diarrhea (e.g., traveler’s diarrhea) because of sunlight, and sunscreens are not always effective.
their activity against many of the causative organisms. Because fluoro-
quinolones vary in their pharmacokinetics and in their spectra, some, Aminoglycosides
but not all, fluoroquinolones are employed for upper and lower respi- The era of the aminoglycosides began in 1943 with the isolation of
ratory tract infections, P. aeruginosa infections, genital diseases caused streptomycin by Waksman and the subsequent development of kana-
by gonococci and Chlamydia, legionnaires disease, and tuberculosis. mycin (1957), gentamicin (1963), tobramycin (1968), amikacin
(1972), and netilmicin (1975).
Therapeutic uses in dentistry
Fluoroquinolones are not indicated for any acute orofacial infections Chemistry
unless dictated by culture and sensitivity tests. Drugs with better anti- Streptomycin is produced by Streptomyces griseus. Other amino-
microbial spectra are readily available. glycosides are elaborated by various species of Streptomyces and
Micromonospora or, in the case of amikacin and netilmicin, are
Adverse effects semisynthetic derivatives of naturally occurring aminoglycosides. As
The major adverse effects associated with fluoroquinolones occur in the name implies, these agents consist of a highly polar amino base
the gastrointestinal tract, CNS, skin, and cartilage. Less common sys- attached by glycosidic linkage to one or more sugars (Fig. 33-14).
tems involved are the cardiovascular, hepatic, and renal systems. The
incidence of each adverse effect may vary among the different drugs Mechanism of action and antibacterial spectrum
because of chemical substitutions on the quinolone nucleus. Aminoglycosides bind irreversibly to the 30S ribosome to interfere
Gastrointestinal adverse reactions include nausea and vomiting, with the reading of the microbial genetic code and to inhibit protein
dyspepsia and heartburn, and abdominal pain. The heavy use of flu- synthesis. Aminoglycosides are generally bactericidal, and their efficacy
oroquinolones has been associated with PMC and diarrhea owing to in several cases can be greatly enhanced by the concomitant use of cell
overgrowth of C. difficile. CNS effects include mild neuropathy (head- wall–inhibiting β-lactams and glycopeptides.
ache, dizziness, malaise, restlessness, bad dreams) possibly caused by The activity of aminoglycosides is primarily directed toward
central γ-aminobutyric acid inhibition or activity at the N-methyl- gram-negative bacilli and mycobacteria (see Table 33-5). The spec-
d-aspartate receptor. Dermatologic toxicity includes rash, pruritus, trum includes gram-negative enteric bacilli and some other gram-neg-
exfoliative dermatitis, Stevens-Johnson syndrome, and phototoxicity ative bacilli (e.g., P. aeruginosa and Proteus). There are, however, some
likely caused by dose-related ultraviolet light activation of reactive oxy- differences among aminoglycosides regarding their efficacy toward
gen from the fluoroquinolones in the skin. specific microorganisms. Gentamicin is the most commonly used ami-
Chondrotoxicity includes arthralgia, joint swelling, tendinitis, and noglycoside, often acting synergistically with ampicillin, penicillin G,
tendon rupture (primarily the Achilles tendon). These disorders are
more likely to occur in men, elderly patients, and patients with con- CH3
comitant corticosteroid therapy, diabetes mellitus, renal failure, other
H3C
musculoskeletal disorders, or involvement in sports activity. These NH
agents are not approved for children younger than 18 years except for
O
ciprofloxacin.
Other possible adverse effects include induction of a prolonged NH2
QT interval leading to torsades de pointes, especially with levofloxa- H2N NH2
cin, gemifloxacin, and moxifloxacin. Fluoroquinolones are associated O

with transient increase in liver enzymes, neutropenia, serum sick- O


HO
ness, allergic vasculitis, and renal crystalluria. Because these antibiot-
ics affect DNA, they have been investigated as possible teratogens. At O CH3
extremely high in vitro doses, fluoroquinolones have induced genotox-
icity in the chromosomal aberration test; however, a clinical study of HO
HN
OH
200 women exposed to ciprofloxacin and norfloxacin during gestation CH3
showed no increase in fetal malformations or musculoskeletal defects. Gentamicin
Exacerbation of myasthenia gravis has been reported. FIG 33-14  Structure of gentamicin.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 481

ceftriaxone, vancomycin, and rifampin. Some original indications for Drug interactions
aminoglycosides have been supplanted by safer extended-spectrum The nephrotoxicity of aminoglycosides is increased by vancomycin and
β-lactams and fluoroquinolones. cephalosporins. Loop diuretics increase auditory toxicity from amino-
glycosides. Aminoglycosides increase the neuromuscular blocking
Bacterial resistance effect of curare-type drugs.
Three resistance mechanisms presently exist for aminoglycosides:
ribosomal mutations (less affinity for the 30S ribosome), reduced Vancomycin
intracellular transport (primarily in staphylococci and pseudo- Vancomycin is a glycopeptide antibiotic, originally isolated from
monads), and, most commonly, plasmid-mediated aminoglyco- Streptomyces orientalis in Borneo in 1956 and introduced into medi-
side-modifying enzymes (acetyltransferases, adenyltransferases, cine in 1958. Vancomycin is a seven-membered peptide chain with two
and phosphotransferases). sugars, vancosamine and glucose. The drug is poorly absorbed from
the gastrointestinal tract and causes severe pain when given intramus-
Absorption, fate, and excretion cularly. It is administered intravenously to treat systemic infections or
Aminoglycosides are poorly absorbed orally and do not penetrate orally to treat PMC. Its elimination half-life is ∼6 hours, and it has a
well into the CNS, bronchial secretions, or certain microbial cells post-antibiotic effect of 1.5 to 3 hours.
(e.g., Rickettsiae, Chlamydiae), but they are effective intracellularly
in the treatment of tuberculosis, plague, brucellosis, and tularemia. Mechanism of action and antibacterial spectrum
Aminoglycosides are classic concentration-dependent antibiotics com- Vancomycin inhibits gram-positive bacterial cell wall synthesis by
monly administered in high parenteral doses repeated after the blood complexing with the d-alanyl-d-alanine portion of the peptide pre-
levels have decreased to a low concentration (peak and trough dos- cursor units to inhibit the transglycosylase reaction in peptidoglycan
ing). Single daily dosing is becoming more common, taking advantage synthesis. This inhibition is at the second stage of bacterial cell wall
of the long post-antibiotic effect of aminoglycosides, a reduction in synthesis before the action of the penicillins at the third stage (see Fig.
cost, and lessening of renal toxicity. The normal elimination half-life 33-5). Vancomycin may also affect cytoplasmic membrane permeabil-
of aminoglycosides is 2 to 3 hours, which can be extended to 24 to ity and RNA synthesis and, as with the β-lactams, it requires active
100 hours in end-stage renal disease. Aminoglycosides are excreted pri- cell replication. Because of its large molecular size, vancomycin cannot
marily by glomerular filtration. traverse the outer cell membrane of gram-negative bacteria.
The activity of vancomycin is almost exclusively against aerobic
General therapeutic uses and anaerobic gram-positive species. Table 33-13 lists several impor­
Parenteral aminoglycosides currently available include amikacin, tant pathogens that are inhibited by the drug.
gentamicin, kanamycin, netilmicin, streptomycin, and tobramycin.
Kanamycin and neomycin are available for oral use (poorly absorbed) Bacterial resistance
for gastrointestinal infections. Aminoglycosides are primarily indicated Vancomycin resistance is caused by an altered peptidoglycan terminus
for infections caused by gram-negative aerobic bacteria, including (d-ala-d-lac instead of the usual d-ala-d-ala), resulting in reduced van-
P. aeruginosa, Serratia, Klebsiella, Enterobacter, and Proteus. Anaerobic comycin binding and failure to prevent cell wall synthesis. Resistance
bacteria are insensitive. Aminoglycosides are often combined with a in vancomycin-intermediate S. aureus and glycopeptide-intermediate
penicillin or cephalosporin for various infections. S. aureus may be due to the production of abnormal peptides (“false
binding sites”) in the cell wall that bind vancomycin and prevent its
Therapeutic uses in dentistry attachment to its receptor or possibly to an increase of peptidoglycan
Aminoglycosides have no uses in orofacial infections unless dictated by resulting in thickened cell walls. A form of resistance is seen in S. pneu­
culture and sensitivity tests. moniae by a unique mutation in the sensor-response system that con-
trols autolysin activity necessary to kill certain bacteria.
Adverse effects
The major adverse effects of aminoglycosides are renal toxicity and
eighth cranial nerve toxicity (auditory and vestibular ototoxicity). TABLE 33-13  Antimicrobial Spectra of
Nephrotoxicity is caused by inhibition of an intracellular lysosomal Systemic Peptide-Containing Antibiotics
phospholipase in the renal proximal tubules, resulting in aminogly- Class Representative Spectrum
coside accumulation and subsequent reduced glomerular filtration,
reduced water and Na+ transport, reduced mitochondrial respiration, Glycopeptide
and reduced renal protein synthesis resulting in renal necrosis. The Vancomycin Staphylococcus including MRSA, enterococci, Streptococ-
incidence of some degree of nephrotoxicity can be 10% to 20%. cus pneumoniae, Streptococcus pyogenes, Peptostrep-
A primary target for toxicity of aminoglycosides is the hair cells of tococcus, VGS, Clostridium difficile, Corynebacterium
the inner ear where the initial loss of the outer hair cells eventually jeikeium, Bacillus cereus, Bacillus subtilis
damages the inner ear cochlear hair cells (type II). Further damage may Lipopeptide
occur to the cochlear sensory epithelium and the spiral ganglion cells Daptomycin MRSA, enterococci, S. Pyogenes, Peptostreptococcus
required for cochlear implants. Vestibular (type I) hair cell damage
Lipoglycopeptides
occurs at the apex of the macula and often earlier than the cochlear hair
cell damage. Initial signs and symptoms are hearing loss at the higher Telavancin MRSA, enterococci, S. Pyogenes, VGS, Peptostreptococ-
frequencies, which increases with dose, duration, and noise exposure. Dalbavancin cus, S. pneumoniae
The incidence of cochlear damage may be 15%. Other adverse reac- Oritavancin
tions associated with aminoglycosides include neuromuscular block- MRSA, Methicillin-resistant Staphylococcus aureus; VGS, viridans
ade of the curare type, rare blood dyscrasias, headache, dizziness, and group streptococci.
urticarial and peripheral neuropathy.
482 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

General therapeutic uses Streptogramins


Vancomycin is used for serious gram-positive infections caused by Quinupristin-dalfopristin, a 30/70 mixture of streptogramins A and
such organisms as methicillin-resistant staphylococci and S. pneu­ B, is approved for intravenous use in the United States.
moniae. It is also useful for non-vancomycin-resistant enterococcal
infections. It is effective in treating enterocolitis caused by C. difficile; Mechanism of action and antibacterial spectrum
however, metronidazole should be used for this situation if possible Quinupristin and dalfopristin bind sequentially to different sites of the
because of the significant risk of promoting vancomycin enterococcal 50S subunit of the 70S ribosome to prevent newly synthesized pep-
resistance. Vancomycin may also be useful in treating multiple antibi- tide chains from extruding from the ribosome, resulting in cell death.
otic-resistant VGS infections (see Table 33-13). Quinupristin-dalfopristin is used to treat life-threatening vanco-
mycin-resistant E. faecium (E. faecalis is resistant) and skin or skin
Therapeutic uses in dentistry structure infections from S. aureus and S. pyogenes. The drug is addi-
Vancomycin has no use in the management of acute or chronic orofacial tionally approved in the United Kingdom for nosocomial pneumonia.
infections unless dictated by laboratory culture and sensitivity tests. Its spectrum closely resembles that of vancomycin and linezolid.

Adverse effects Bacterial resistance


Major adverse drug reactions associated with vancomycin include Microbial resistance occurs by three mechanisms: decreased ribo-
transient or permanent ototoxicity, hypotension, reversible neutrope- somal binding by methylation of an adenine residue, drug efflux,
nia, renal toxicity, skin rash, and red man syndrome. The auditory and enzymatic inactivation. This resistance belongs to the MLSB
toxicity of vancomycin is dose-dependent and can be exacerbated by (Macrolide-Lincosamide-Streptogramin B) type, potentially confer-
the combination with aminoglycosides. Red man syndrome results ring cross-resistance to all these antibiotics.
from the direct histamine release from mast cells manifesting as pru-
ritus; erythematous rash of the head, neck, face, and upper torso; and General therapeutic uses
hypotension mimicking anaphylactic shock. Rapid infusion makes it Quinupristin-dalfopristin should be reserved for life-threatening and
worse. This glycopeptide-induced anaphylactoid reaction may occur multiple antibiotic-resistant infections from E. faecium, staphylococci,
with the first drug exposure, is tachyphylactic in nature, and can be and some streptococci encountered primarily in the hospital.
reduced significantly by the slow infusion of vancomycin over a 1-hour
period and premedication with antihistamine drugs. To reduce the Adverse effects
development of resistant bacteria, vancomycin use is contraindicated Adverse effects associated with streptogramins include possible severe
or discouraged for routine surgical prophylaxis. arthralgias and myalgias, elevation in conjugated serum bilirubin, and
significant inhibition of the liver microsomal CYP3A4 drug metabo-
Drug interactions lizing system.
Vancomycin-induced nephrotoxicity or ototoxicity is increased with
the concomitant use of aminoglycosides, and neuromuscular blockade Drug interactions
with curare-like agents is enhanced by vancomycin. Streptogramins decrease the liver metabolism of Ca2+ channel block-
ers, immunosuppressant drugs, corticosteroids, several anticancer
Fidaxomicin agents, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors
Fidaxomicin is a macrolide antibiotic whose spectrum is narrow and (statins), HIV protease inhibitors, quinidine, nonsedating antihista-
limited to gram-positive aerobes and anaerobes. It inhibits RNA poly- mines, sildenafil, opioids, and benzodiazepines.
merase and is bactericidal against C. difficile. It is given orally to treat
colitis due to C. difficile. Systemic absorption is minimal when given Oxazolidinones
orally. Two oxazolidinones, linezolid and tedizolid, are totally synthetic anti-
microbial drugs.
Daptomycin
Daptomycin is a cyclic lipopeptide bacteriocidal antibiotic. Its causes Mechanism of action and antibacterial spectrum
depolarization of sensitive gram-positive bacteria after binding to The oxazolidinones have a unique mechanism of action by binding
their cell membranes. Its spectrum includes MRSA, enterococci, S. to the 50S ribosome subunit near the interface with the 30S subunit
Pyogenes, and Peptostreptococcus (see Table 33-13). It is adminis- to prevent the initiation complex required for bacterial translation.
tered intravenously and is excreted unchanged by the kidneys. Adverse This unique mechanism may possibly limit cross-resistance with other
effects include skeletal muscle damage and peripheral neuropathy. antibiotics. Linezolid is approved in the United States for the manage-
ment of vancomycin-resistant E. faecium; nosocomial and commu-
Lipoglycopeptides nity-acquired pneumonia from S. aureus and penicillin-sensitive S.
Telavancin, dalbavancin, and oritavancin are intravenous drugs that pneumoniae; and complicated skin and skin structure infections from
inhibit cell wall synthesis in gram-positive cocci, similar to vanco- MRSA, MSSA, methicillin-resistant CoNS, S. pyogenes, and S. agalac­
mycin. In addition, telavancin has the additional effect of causing tiae. Tedizolid has similar indications.
cell depolarization by acting on the cell membrane. The spectrum
of these drugs includes MRSA, enterococci, S. Pyogenes, VGS, Bacterial resistance
Peptostreptococcus, and S. pneumoniae (see Table 33-13). The half- Microbial resistance to linezolid has been detected in isolated cultures
lives are telavancin, ∼8 hours; dalbavancin, ∼6 to 11 days; and orita- of vancomycin-resistant enterococci, E. coli, and laboratory strains of
vancin, ∼10 days. Telavancin causes taste disturbances and is linked to S. aureus. More recent reports of clinical isolates of E. faecium and S.
QT prolongation and nephrotoxicity. Dalbavancin and oritavancin are aureus resistant to linezolid are disconcerting because the drug has
associated with GI disturbances and hepatotoxicity. The drugs are used been available only for a short time. The mechanism of resistance
to treat complicated skin and soft tissue infections. seems to be a 62,576T mutation in the gene encoding the central loop
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 483

domain of 23S rRNA. Tedizolid may be active against certain strains of significance of this drug effect is as yet unknown. GI disturbances,
bacteria resistant to linezolid. tachycardia, and hypertension are among the adverse effects associated
with tedizolid.
Absorption, fate, and excretion
Linezolid, with a near 100% oral bioavailability, produces peak blood Sulfonamides
levels at 1 to 2 hours and can be administered parenterally as well. It The era of effective and safe systemic antimicrobial therapy began in
has an elimination half-life of 4.4 to 5.5 hours and a post-antibiotic 1932 with the discovery by Domagk that a dye (Prontosil) protected
effect of 0.6 to 1.4 hours. Tedizolid has a half-life of ∼12 hours. laboratory animals from streptococcal infections. Domagk deter-
mined that the active antibacterial portion of Prontosil was sulfanil-
Adverse effects amide, which was subsequently first used in the United States in 1935.
Approximately 2% to 3% of patients receiving linezolid experience Trimethoprim was introduced in 1968 as a synergistic agent with
nausea and vomiting, diarrhea, headache, tongue discoloration, taste sulfonamides, and the combination of sulfamethoxazole/trimetho-
alteration, fungal superinfections, or very rarely PMC. The most seri- prim is the most commonly used sulfonamide preparation today.
ous adverse reaction is myelosuppression (anemia, leukopenia, pan- The discovery of Prontosil by Domagk ranks with the discovery of the
cytopenia, thrombocytopenia), which may occur in an average of 2.4% anesthetic properties of nitrous oxide by Wells, the work on penicillin
of patients. Linezolid requires weekly blood monitoring tests. The by the Oxford group, and the discovery by Jenner of vaccinations as
safety of the drug has not been established beyond 28 days of use. It among the greatest of all medical discoveries.
is classified as an FDA pregnancy category C drug. Adverse effects of
tedizolid are similar to those of linezolid. Chemistry
All the sulfonamides are derivatives of p-aminobenzenesulfonamide.
Drug interactions Sulfonamides are weak acids with limited water solubility, particularly
Linezolid is a weak monoamine oxidase inhibitor and should be in solutions of low pH. This property may present problems for the
used with caution with drugs that release catecholamines and foods excretion of these drugs in acidic urine.
containing tyramine. Linezolid may precipitate serotonin syndrome
(confusion, agitation, seizures, hypertension, tachycardia, sweat- Mechanism of action and antibacterial spectrum
ing, myoclonus, muscle rigidity, trismus, death), but the clinical Sulfonamides and trimethoprim interfere with the microbial synthe-
sis of folic acid necessary for life in some microorganisms (Fig. 33-15).
Mammals acquire folic acid from their diet. Sulfonamides competi-
tively inhibit the incorporation of PABA into dihydropteroic acid, a
Dihydropteroate synthase
precursor of dihydrofolate, because the sulfas have a greater affinity
for dihydropteroate synthetase than PABA. Trimethoprim inhibits
PABA + pteridine Dihydropteroic acid bacterial dihydrofolate reductase with 50,000 to 100,000 times greater
affinity for the bacterial than the human enzyme; this blocks the con-
version of dihydrofolic acid to tetrahydrofolic acid, which is important
Sulfonamides in the synthesis of purines, and DNA. Sulfonamides and trimethoprim
inhibit successive steps in the synthesis of folic acid and eventually bac-
terial nucleotides and DNA.
Dihydrofolate reductase
Dihydrofolic acid
The trimethoprim/sulfamethoxazole combination is commonly
used for respiratory, urinary, and gastrointestinal infections. This
combination is used for indications indicated in Table 33-14.
Tetrahydrofolic acid Trimethoprim
Bacterial resistance
Transferable microbial resistance to sulfonamides and trimethoprim
occurs by three principal mechanisms: increased cell permeability bar-
Purine and pyrimidine synthesis riers and efflux proteins, decreased sensitivity or alterations in target
FIG 33-15 Sites of inhibition of folic acid synthesis by sulfonamides enzymes (dihydropteroate synthase and dihydrofolate reductase), and
and trimethoprim. Note the effect of the two drugs on this common the acquisition of new target enzymes. Resistance genes for trimetho-
pathway and the effect on purine and pyrimidine synthesis. PABA, prim-sulfamethoxazole are carried on transposable elements along
p-Aminobenzoic acid. with the resistance genes for other antibacterial drugs.

TABLE 33-14  Characteristics of Representative Sulfonamides*


Drug Half-Life (hrs) Route Spectrum
Sulfisoxazole ∼6 Oral Escherichia coli, Shigella, Yersinia enterocolitica, Burkholderia cepacia, Stenotrophomonas maltophilia, Nocar-
Trimethoprim/­ ∼11/∼11 Oral dia, Moraxella catarrhalis, MRSA, Streptococcus pneumoniae, Listeria monocytogenes, Bartonella henselae,
sulfamethoxazole Brucella, Vibrio cholerae, Mycobacterium marinum, Burkholderia pseudomallei
Mafenide — Topical (burns) —
Silver sulfadiazine — Topical (burns) —
Nocardia is an indication for sulfisoxazole alone.
*Antimicrobial spectrum corresponds to trimethoprim/sulfamethoxazole, except where noted.
484 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

Absorption, fate, and excretion and Rickettsiae. Common pathogens sensitive to chloramphenicol
Sulfonamides are classified as short-acting, medium-acting, or long- include Salmonella typhi, other Salmonella species, S. pneumoniae, H.
acting. Short-acting to medium-acting agents include sulfisoxazole, sul- influenzae, and N. meningitidis. The drug is an alternate drug for seri-
famethoxazole, sulfamethizole, and sulfadiazine. Sulfadoxine is a long- ous infections such as bacterial meningitis and rickettsial diseases such
acting agent with an elimination half-life of 100 to 230 hours (it is as Rocky Mountain spotted fever.
used in combination with pyrimethamine to treat malaria caused by The most significant adverse reactions associated with chloramphen-
Plasmodium falciparum). Various other sulfonamide preparations icol are reversible and irreversible bone marrow depression seen with
include preparations used topically for burns (silver sulfadiazine, oral, parenteral, and even topical use. (The drug is absorbed well by any
mafenide), vaginal preparations, ophthalmic preparations (sulfacet- route.) The reversible type is dose-related and possibly caused by inhi-
amide), and drugs for the management of ulcerative colitis (salicylazo- bition of mitochondrial protein synthesis resulting in anemia, leukope-
sulfapyridine or sulfasalazine) given orally for local gastrointestinal nia, or thrombocytopenia. “Idiosyncratic” bone marrow aplasia is not
effects. Oral sulfonamides are 70% to 100% bioavailable (except for dose-related; may begin weeks or months after the drug is stopped; and
sulfasalazine) with a large volume of distribution and ready penetra- is manifested by an often fatal aplastic anemia, the incidence of which
tion into all fluids and tissues in the body including the CNS. The seems to be 1 in 24,500 to 1 in 40,800 patients receiving chloramphenicol
drugs are metabolized by acetylation and conjugation in the liver and by any route of administration. This incidence is 13 times greater than
excreted by glomerular filtration. the spontaneous random occurrence of aplastic anemia in the general
population. Topical use is associated with a risk of 3 cases in 440,000
General therapeutic uses uses. The cause of this idiosyncratic aplastic anemia is unknown, but it
The primary clinical uses of a sulfonamide alone or trimethoprim/sul- may be due to a genetically determined liver metabolite.
famethoxazole are genitourinary tract infections, otitis media, acute The “gray baby syndrome” associated with chloramphenicol is
bronchitis, community-acquired pneumonia, and traveler’s diar- caused by toxicity resulting from the inability of the immature liver of
rhea from enterotoxigenic bacteria. Two topical sulfonamides, silver neonates to detoxify the drug by conjugation. The signs and symptoms
sulfadiazine and mafenide, are also used to treat burns. Other sulfon- include abdominal distress, cyanosis, vomiting, circulatory collapse,
amides are used for ophthalmic and local gastrointestinal indications. and possibly death. There are no indications for chloramphenicol
There are no indications for sulfonamides and trimethoprim in the in the management of orofacial infections. The drug is rarely used
management of orofacial infections. because of its major adverse effects, especially bone marrow aplasia.

Adverse effects Urinary Antiseptics


Approximately 8% of individuals receiving sulfonamides and tri- Nitrofurantoin
methoprim have some form of adverse reaction, such as nausea and Nitrofurantoin is prepared in various suspension forms and, as
vomiting, blood dyscrasias, and crystalluria (precipitation in the with all urinary antiseptics, has limited bioavailability, low volumes
urine more likely with less soluble preparations such as sulfadiazine). of distribution, and high urinary excretion rates. Its mechanism of
Three to five percent of patients experience allergy in any of its forms action is unknown but may involve inhibition of DNA and pro-
from skin rash and pruritus to major skin eruptions (Stevens-Johnson tein synthesis after its enzymatic activation in the bacterial cell. Its
syndrome, epidermal necrolysis, exfoliative dermatitis, photosensitiv- antibacterial spectrum includes E. coli, Citrobacter, Staphylococcus
ity) to anaphylaxis. Stevens-Johnson syndrome is much more likely to saprophyticus, E. faecalis, group B streptococci, K. pneumoniae,
occur with long-acting sulfonamides, which are not available in the and Enterobacter, with inherent resistance in Proteus, Providencia,
United States. Of patients with AIDS, 70% have some form of skin rash Morganella, Serratia, Acinetobacter, and P. aeruginosa. Adverse drug
and fever from sulfonamides and trimethoprim. reactions include severe gastrointestinal upset (nausea and vomiting,
anorexia, cramping), hepatitis, pneumonitis, peripheral neuropa-
Drug interactions thy, and bone marrow suppression. Pulmonary pneumonitis may
Trimethoprim and sulfamethoxazole work together synergistically in their be acute, subacute, or chronic with an incidence for the acute form
antimicrobial effect, increasing the activity and spectrum of the other. of 1 in 100,000 users. Hemolytic anemia may occur in individuals
Trimethoprim/sulfamethoxazole increases the clinical activity of oral anti- deficient in glucose-6-phosphate dehydrogenase. Nitrofurantoin and
coagulants, thiopental, methotrexate, hydantoins, and sulfonylureas, and the other agents mentioned subsequently are indicated for uncompli-
it decreases the activity of cyclosporine. Sulfonamides are displaced from cated urinary tract infections and cystitis.
plasma protein by aspirin, other NSAIDs, and probenecid. PABA (present
in some health foods) competes against the effect of sulfonamides. Fosfomycin
Fosfomycin is a broad-spectrum bactericidal drug that is converted in
Contraindications the blood to the free acid form of fosfomycin. Its mechanism of action
Contraindications for the use of sulfonamides include allergy to sul- is to inactivate enolpyruvyl transferase responsible for the conden-
fonamides and other related drugs, such as sulfonylureas and thia- sation of uridine diphosphate-N-acetylglucosamine with p-enolpyru-
zide, loop, and carbonic anhydrase inhibitor diuretics. Health foods vate, one of the initial steps in microbial cell wall synthesis (see Fig.
and possibly sunscreens containing PABA are contraindicated because 33-5). The antimicrobial spectrum for fosfomycin includes E. coli, E.
PABA competes against the sulfonamide. faecalis, Citrobacter, Enterobacter, K. pneumoniae, P. mirabilis, and S.
marcescens. Adverse reactions are mild and include diarrhea, vaginitis,
Chloramphenicol rash, and headache. Use of fosfomycin is commonly restricted to only
Chloramphenicol (Chloromycetin) is a broad-spectrum antibiotic a single dose because of rapid microbial resistance.
isolated in 1949 from Streptomyces venezuelae. It is bacteriostatic and
inhibits bacterial protein synthesis by reversible binding to the pepti- Methenamine
dyl transferase component of the 50S ribosomal subunit. The hydrolysis of methenamine, below pH 5.5, results in the liberation
Chloramphenicol is a broad-spectrum antibiotic whose spectrum of formaldehyde as its component. This results in the denaturation of
includes several gram-positive and gram-negative bacteria, spirochetes, proteins. Methenamine, as the hippurate or mandelate salt to acidify
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 485

the urine, has a broad spectrum of activity against E. coli, staphylo- Isoniazid is well absorbed after either oral or parenteral adminis-
cocci, and enterococci, with significant resistance in P. vulgaris and P. tration, but the oral route is preferred for reasons of convenience and
aeruginosa. It is used as suppressive or prophylactic therapy in chronic maximum therapeutic effect. The drug is well distributed into all body
urinary tract infections. Adverse reactions include pruritus, urticaria, fluids, including the caseous material of the tubercle-infected foci.
nausea and vomiting, cramping, headache, dizziness, proteinuria, Isoniazid is mainly metabolized in the liver and excreted in the urine
hematuria, and precipitation of urate crystals in the urine. as metabolites. Genetic differences in the rate of biotransformation are
seen, but these seem to have little effect on therapeutic efficacy. The
Nalidixic acid plasma half-life is prolonged in patients with hepatic dysfunction.
This quinolone antibiotic is used exclusively for the management One important adverse reaction with isoniazid is peripheral neu-
of urinary tract infections from gram-negative microorganisms. ritis caused by an isoniazid-induced increase in the excretion of pyr-
(See Table 33-12.) Its mechanism of action is the same as the fluo- idoxine. This adverse effect is more common in slow acetylators.
roquinolones: inhibition of DNA gyrase and topoisomerase IV. The This reaction and other symptoms of pyridoxine deficiency can be
major adverse effects are CNS toxicity (dizziness, weakness, headache, prevented by prophylactic administration of vitamin B6 (15 to 50 mg
papilledema, and rare seizures and psychosis), blood dyscrasias, pho- daily). Other adverse effects include allergic reactions (fever, rashes,
tosensitivity, and hemolytic anemia in glucose-6-phosphate dehydro- hepatitis), fatal hepatic necrosis (rarely), xerostomia, epigastric dis-
genase–deficient individuals. tress, hematologic reactions, and convulsions in seizure-prone patients
(although administration of isoniazid to patients taking phenytoin
Drugs Used to Treat Tuberculosis has not been problematic except for the potential of pharmacokinetic
Successful treatment of tuberculosis caused by M. tuberculosis became effects on phenytoin metabolism). A nonallergic hepatitis of some
possible only with the advent of chemotherapeutic agents. Multidrug- severity has also been reported, and subsequent studies have shown
resistant strains of M. tuberculosis have arisen, especially among that the incidence of hepatic damage increases with age and in individ-
patients with HIV/AIDS. Because of the rapid development of antimi- uals who regularly drink alcohol.
crobial resistance in strains of M. tuberculosis, a combination of agents Isoniazid is also effective prophylaxis against tuberculosis and
is always employed for treatment. The primary (first-line) antituber- approved for single-drug therapy for prophylaxis. It is also the most
culosis drugs are isoniazid, rifampin, pyrazinamide, ethambutol, important drug used in tuberculosis therapy for reasons of effective-
and streptomycin. (Rifabutin or rifapentine can be used to substitute ness, expense, convenience of administration, and relative safety.
for rifampin if needed.) (Depending on the reference, streptomycin is
classified either as a first-line of second-line drug.) For recurrent infec- Rifampin
tions or cases that exhibit microbial resistance, second-line drugs are Rifampin is a semisynthetic derivative of one of the rifamycins, a group
available, including ethionamide, cycloserine, amikacin, kanamycin, of macrocyclic antibiotics produced by Streptomyces mediterranei.
capreomycin, levofloxacin, moxifloxacin, para-aminosalicylic acid, Rifampin is effective against numerous gram-positive and gram-neg-
and bedaquiline. These agents are generally less active and often more ative bacteria in addition to M. tuberculosis and most other species of
toxic than the primary drugs. These drugs are used in combination for Mycobacterium. Its mechanism of action involves inhibition of DNA-
active tuberculosis that is drug-resistant. dependent RNA polymerase. Mammalian RNA polymerase does not
Until the results of sensitivity tests dictate the regimen, tuber- bind the drug, and RNA synthesis in host cells is unaffected. Resistance
culosis therapy should begin with four drugs: isoniazid, rifampin, can develop rapidly to rifampin, frequently in a single step, by alter-
pyrazinamide, and ethambutol (or streptomycin) for 2 months, fol- ation of the target enzyme.
lowed by 4 (or 7) months of isoniazid and rifampin. After test results, Rifampin is generally well absorbed from the gastrointestinal tract
typical therapy consists of isoniazid, rifampin, and pyrazinamide for after oral administration. The drug is distributed throughout the body
2 months followed by isoniazid and rifampin for 4 months, or isoniazid and imparts an orange-red color to the urine, saliva, sweat, tears, spu-
and rifampin for 7 months, the length depending on whether there is tum, and feces. It is secreted in the bile and undergoes enterohepatic
a pulmonary cavity on the chest X-ray at presentation and/or a posi- recirculation, prolonging its half-life. Elimination occurs by hepatic
tive sputum culture observed after 2 months of therapy. Other treat- deacetylation and excretion in the urine and feces.
ment times are recommended for TB meningitis and TB osteomyelitis. Rifampin may be useful in prophylaxis of tuberculosis in contacts
Other drug options listed in Table 33-5 are available in multidrug- of patients infected with isoniazid-resistant organisms. The drug has
resistant tuberculosis. The pharmacologic features of isoniazid, rifam- proven effective in certain diseases refractory to conventional therapy,
pin, pyrazinamide, and ethambutol are described here. Streptomycin such as rifampin in combination as an option in treating resistant S.
and other aminoglycoside antibiotics have been previously discussed. pneumoniae and methicillin-resistant staphylococci.
The incidence of adverse reactions to rifampin is low (4%), and
Isoniazid the most common is liver toxicity. Gastrointestinal disturbances, sup-
Isoniazid, the name of which derives from its chemical designation pression of T-lymphocyte function, neurologic disorders, and various
of isonicotinic acid hydrazide, is the most important drug for the allergic reactions, including soreness of the mouth and tongue, have
treatment and prophylaxis of tuberculosis. Its spectrum of activity is been reported. Decreased effectiveness of oral anticoagulants, oral
limited, however, to M. tuberculosis and one species of atypical myco- contraceptives, estrogens, and glucocorticoids have occurred with
bacteria, Mycobacterium kansasii. concomitant administration of rifampin because rifampin induces
Isoniazid inhibits the synthesis of mycolic acids, unique and nec- liver microsomal enzymes. Oral contraceptives are not likely to be
essary components of the cell wall of mycobacteria. The drug is bacteri- effective in a patient taking rifampin.
cidal to actively growing tubercle bacilli but not to dormant organisms. If rifampin is used sporadically, a flulike syndrome (possibly
Resistance to isoniazid occurs by spontaneous mutation of the bac- immune related) may develop, sometimes leading to renal failure,
terial chromosome at a rate of ∼1 in 106 divisions. Most established hepatorenal syndrome, hemolysis, and thrombocytopenia. The drug
infections can be expected to harbor at least several resistant bacteria. should be taken according to a prescribed regimen. Because rifampin
There is no cross-resistance between isoniazid and other antitubercu- can cause a reddish-orange color in body fluids, staining of soft contact
losis drugs except ethionamide. lenses may occur.
486 CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy

Rifabutin Drugs Used to Treat Leprosy


Rifabutin is chemically similar to rifampin and acts by a similar mecha- Although leprosy is rarely seen in the United States, the World Health
nism. Rifabutin is not as potent an inducer of cytochrome P450 enzymes Organization estimates that 12 million cases exist throughout the
as rifampin and offers the advantage of not interacting with other drugs world. Leprosy is a bacterial disease caused by the tubercle bacil-
to the extent that rifampin does. It is useful in treating patients who have lus Mycobacterium leprae. Treatment may be 2 to 4 years or extend
HIV/AIDS because it has less interaction with protease inhibitors and throughout the patient’s life, depending on the severity and type of
non-nucleosidase reverse transcriptase inhibitors. Adverse effects include disease.
uveitis and neutropenia but are otherwise similar to those of rifampin. Dapsone is the major drug used in the treatment of leprosy. It
belongs to a group of drugs called sulfones, which are chemical rela-
Rifapentine tives of sulfonamides. Dapsone is bacteriostatic against M. leprae, with
Rifapentine is a long-acting rifampin-type drug that has a similar a mechanism of action similar to that of sulfonamides. Dapsone is used
mechanism of action and similar adverse effects. It can be used twice orally. Other drugs, normally used in combination with dapsone, are
weekly for initial treatment and once weekly during the continuation rifampin and clofazimine. Clarithromycin, minocycline, and ofloxacin
phase of treatment. may also be effective.

Pyrazinamide Topical Antibiotics


Pyrazinamide is the pyrazine analogue of nicotinamide. It is converted Bacitracin
to pyrazinoic acid by bacterial pyrazinamidase. Pyrazinoic acid inhib- Bacitracin is a polypeptide antibiotic derived from B. subtilis that
its mycolic acid synthesis, and it disrupts cell wall function. It had functions to block cell wall formation by interfering with the dephos-
widespread use in the 1960s but proved to be hepatotoxic in the doses phorylation of the lipid compound that carries peptidoglycans to the
used and was relegated to secondary status after the development of growing microbial cell wall (see Fig. 33-5). The antibacterial spectrum
isoniazid and rifampin. More recently, pyrazinamide in reduced dos- of bacitracin is gram positive and includes staphylococci, streptococci,
age has reemerged as the third most important anti-tuberculosis agent. Corynebacterium, and Clostridium, with rare resistance seen in staphy-
Resistance to the drug in M. tuberculosis infection is associated with the lococci. Bacitracin is too toxic to be used parenterally but is well toler-
loss of pyrazinamidase activity. ated topically. Bacitracin is commonly combined with neomycin and
Pyrazinamide is well absorbed after oral administration and is dis- polymyxin B in over-the-counter topical antibiotic preparations, but
tributed throughout the body. It is metabolized primarily in the liver evidence for efficacy is limited.
and excreted largely in the urine.
Pyrazinamide is administered with other anti-tuberculosis drugs to Neomycin
decrease the duration of therapy required to effect a cure of uncom- Neomycin is an aminoglycoside derived from Streptomyces fradiae
plicated tuberculosis. Hepatotoxicity is the most common adverse and binds to the 30S ribosomal subunit to inactivate bacterial DNA
effect, but this has been less evident with the lower dosages currently polymerase and causes misreading of the genetic code to produce
used. Other toxic effects associated with current regimens are relatively lethal proteins. Neomycin has a wide antibacterial spectrum against
benign or infrequent. Gastrointestinal disturbances, arthralgias, fever, gram-positive and gram-negative bacteria, but it is ineffective against
and rash have been noted. Pyrazinamide may cause hyperuricemia, streptococci and P. aeruginosa.
and the drug represents a risk in patients with gout.
Polymyxin B
Ethambutol Polymyxin B was isolated from Bacillus polymyxa and functions as a
Ethambutol is a synthetic agent that inhibits arabinosyl transferases, cationic detergent to disrupt the microbial cell membrane, causing a
which are important in cell wall synthesis of sensitive mycobacteria. It is leak in cell constituents. Its spectrum is gram negative, and it is par-
active against M. tuberculosis and some other mycobacteria such as M. ticularly useful against P. aeruginosa. The drug is not used parenterally
kansasii. Other bacteria are not affected by the drug. Ethambutol is tuber- because it commonly induces paresthesias, ataxia, and slurred speech.
culostatic, and resistance develops, although slowly, if it is used alone.
Ethambutol is given orally because of good absorption from the Mupirocin
gastrointestinal tract. Distribution into various body compartments Mupirocin has a unique chemical structure composed of a short fatty
is adequate. The major route of excretion of ethambutol is by renal acid chain linked to monic acid; it inhibits bacterial RNA and protein
tubular secretion and glomerular filtration, with the drug appearing synthesis by binding to isoleucyl-tRNA synthetase to prevent incorpo-
in the urine mostly as unchanged drug and as two metabolites. Dosage ration of isoleucine into bacterial proteins. The antimicrobial spectrum
adjustment is required in the presence of renal impairment. for mupirocin includes staphylococci (MRSA, MSSA), streptococci,
Adverse reactions to ethambutol are infrequent, the most notable anaerobes, and Enterobacteriaceae. The primary use of mupirocin is
being optic neuritis, with symptoms of decreased visual acuity and loss of as a topical application for skin infections, such as impetigo due to S.
the ability to perceive the color green. Other adverse effects include gastro- aureus or S. pyogenes. Mupirocin is also used to reduce or eliminate
intestinal upset; peripheral neuritis; allergic reactions, usually appearing as nasal carriage of staphylococci, particularly MRSA. Its widespread use
skin rashes or drug fever; and increased retention of uric acid. is associated with an increase in the reinfection rate due to resistance
development or reinfection from other body areas.
Second-line drugs
A number of second-line drugs are used to treat tuberculosis. These Retapamulin
are useful in cases of resistance to first-line drugs and include ethion- Retapamulin belongs to a group of drugs called the pleuromutilins.
amide, capreomycin, kanamycin, amikacin, para-aminosalicylic acid The drug inhibits protein synthesis by binding to the 50S ribosomal
(whose mechanism is similar to that of the sulfonamides), cycloserine subunit. Retapamulin inhibits S. pyogenes and S. aureus (methicillin
(which inhibits cell wall synthesis), levofloxacin, moxifloxacin, and susceptible). It also has activity against some gram-negative bacteria
other fluoroquinolones. and many anaerobes. It is used to treat impetigo.
CHAPTER 33  Pharmacology of Specific Drug Groups: Antibiotic Therapy 487

ANTIBACTERIAL ANTIBIOTICS* CASE DISCUSSION


Nonproprietary (Generic) Name Proprietary (Trade) Name The most likely diagnosis based on the age and circumstances of the patient is
Aminoglycosides acute necrotizing ulcerative gingivitis (ANUG). The patient’s chills and temperature
Amikacin Amikin indicate that she has signs of a systemic infection, most likely due to the spread of
Gentamicin Garamycin, Jenamicin the oral infection. You do two things at the first appointment: (1) debridement and
Kanamycin Kantrex (2) prescribe amoxicillin 500 mg t.i.d. for 7 to 10 days. Although ANUG should be
Neomycin Mycifradin treated by local debridement, you reason that the antibacterial drug is needed to
Netilmicin Netromycin reduce the systemic involvement. Amoxicillin is an extended-spectrum penicillin.
Paromomycin Humatin When you begin debridement, the patient may complain of pain. As a result
Streptomycin — you may use mild ultrasonic scaling to remove some of the supragingival
Tobramycin Nebcin plaque in the first appointment. Before dismissing the patient, you give her
instructions on good oral hygiene, proper diet, and, if the patient is a smoker,
Antituberculosis Drugs (Not Included Elsewhere in This List) to enter a smoking cessation program. You instruct the patient to call in three
Aminosalicylate sodium Tubasal days to check on progress. You find that the patient is much better in three
Bedaquiline Sirturo days and then schedule a second appointment to complete the scaling and
Capreomycin Capastat Sulfate removal of plaque.
Cycloserine Seromycin Adverse effects of amoxicillin or other penicillins include allergies that can
Ethambutol Myambutol range from mild to anaphylactic reactions. Amoxicillin is an extended-spectrum
Ethionamide Trecator antibiotic. This could lead to a superinfection if given for several days. Amoxi-
Isoniazid Nydrazid cillin is usually well tolerated.
Pyrazinamide —
Rifabutin Mycobutin
Rifampin Rifadin, Rimactane
Rifapentine Priftin
GENERAL REFERENCES
1. Abramowicz M, Zuccotti G, editors: Handbook of antimicrobial drugs:
Topical Antibiotics
drugs for bacterial infections. The Medical Letter, ed 20, New Rochelle,
Bacitracin Baciguent NY, 2015.
Mupirocin Bactroban 2. Albert RK, Schuller JL: Macrolide antibiotics and the risk of cardiac
Neomycin Myciguent arrhythmias, Am J Respir Crit Care Med 189:1173–1180, 2014.
Polymyxin B Aerosporin 3. Bennett PM: Plasmid encoded antibiotic resistance: acquisition and transfer
Retapamulin Altabax of antibiotic resistance genes in bacteria, Br J Pharmacol 153:S347–S357, 2008.
Bacitracin with neomycin and polymyxin B Neosporin 4. Bennett JE, Dolin R, Blaser MJ, editors: Mandell, Douglas, and Bennett’s
principles and practice of infectious diseases, ed 8, New York, Saunders,
Miscellaneous Agents 2015, Elsevier.
Chloramphenicol Chloromycetin 5. Cho H, Uehara T, Bernhardt TG: Beta-lactam antibiotics induce a lethal
Clofazimine Lamprene malfunctioning of the bacterial cell wall, Cell 159:1300–1311, 2014.
Colistimethate Coly-Mycin M 6. Drugs for bacterial infections, Med Letter 11(131):65–74, 2013.
Colistin In Coly-Mycin S 7. Drugs for tuberculosis, Med Letter 10(116):29–36, 2012.
Dalbavancin Dalvance 8. Leffler DA, Lamont JT: Clostridium difficile infection, N Engl J Med
372:1539–1548, 2015.
Dapsone —
9. Macy E: Penicillin and beta-lactam allergy: epidemiology and diagnosis,
Daptomycin Cubicin
Curr Allergy Asthma Rep 14:476, 2014 (published online).
Fidaxomicin Dificid 10. Salyers AA, Whitt DD: Bacterial pathogenesis: a molecular approach, ed 2,
Fosfomycin Monurol Washington, DC, 2002, ASM Press.
Lincomycin Lincocin
Linezolid Zyvox
Methenamine Hiprex, Mandelamine, Urex
Metronidazole Flagyl
Nitrofurantoin Furadantin, Macrodantin
Oritavancin Orbactiv
Quinupristin/dalfopristin Synercid
Tedizolid Sivextro
Telavancin Vibativ
Telithromycin Ketek
Tigecycline Tygacil
Troleandomycin TAO
Vancomycin Vancocin

*Agents not shown here are listed in various tables throughout this
chapter.

You might also like