You are on page 1of 20

Cephalosporins

21  William A. Craig and David R. Andes

Although the discovery of the cephalosporin antibiotic class was Modifications at R2 often alter the pharmacology of the compound.
reported in 1945, it took almost 2 decades for this class to achieve Changes in the R2 constituent may influence the ability of the com-
clinical utility. Giuseppe Brotzu is widely credited for discovery of the pound to reach certain infection sites, such as the central nervous
broad-spectrum inhibitory effects of sewage outflow in Sardinia, Italy.1 system, or may simply prolong the elimination half-life of the drug.
Professor Brotzu subsequently isolated the mold Cephalosporin acre- The predominant changes at R1 (position C7) include the addition
monium (now Acremonium chrysogenum) and demonstrated antimi- of an acyl side chain and the substitution of the hydrogen with
crobial activity of culture filtrates against both gram-positive and a methoxy group.5 The R1 methoxy substitution led to the develop-
gram-negative bacteria. He also demonstrated the in vivo activity of ment of the cephamycin group of compounds, including cefoxitin,
these culture filtrates in animal infection models and in several patients. cefmetazole, and cefotetan. This alteration enhanced resistance
The filtrate was administered both locally by injection into skin to β-lactamase produced by gram-negative anaerobic and aerobic
abscesses and systemically for the treatment of brucellosis and typhoid bacteria.5,6 However, these compounds have lower affinity for the
fever. penicillin-binding protein (PBP) target in gram-positive bacteria.7 The
A decade after the initial discovery, the cephalosporin substances cephamycin group is structurally related to the cephalosporins but
were isolated and identified as fermentation products of the mold.2 originated as a metabolite from Streptomyces lactamdurans.6 The basic
Investigators at Oxford, including Florey and Abraham, systematically building block of the cephamycin group is cephamycin C. Hydrolysis
studied the physical, chemical, and structural characteristics of cepha- of cephamycin C produces the 7-ACA nucleus.
losporins, as they had for the penicillin class a decade earlier. Three Many modifications of the acyl side chain have been undertaken.
substances—cephalosporin P, N, and C—were identified. Each of the The first compounds resulting from addition of a thienyl ring or a
products possessed antimicrobial activity but only cephalosporin C tetrazole structure at R1 included the first-generation cephalosporins—
demonstrated activity against both gram-negative and gram-positive cephalothin, cephaloridine, and cefazolin (Fig. 21-2). The simple sub-
bacteria. In addition, it had advantageous stability in the presence of stitution of an aminobenzyl group in the C7 position is important for
acid and penicillinases.2 Cephalosporin C became the foundation of oral absorption of the cephalosporins.5 Cephalexin, cephradine, cefa-
subsequent drug development. clor, cefprozil, and loracarbef all have this structure or a closely related
The first cephalosporin pharmaceutical, cephalothin, was intro- one (Fig. 21-3). Absorption of later-generation cephalosporins is
duced for clinical use in 1964. There are more than 20 cephalosporin enhanced by the production of ester formulations. Axetil, proxetil, or
antibiotics in use today (17 in the United States). The cephalosporin pivoxil esters of cefuroxime, cefpodoxime, and cefditoren are currently
class is among the most widely prescribed antimicrobial classes because available.
of its broad spectrum of activity, low toxicity, ease of administration, Most of the chemical modifications in cephalosporin development
and favorable pharmacokinetic profile. that have resulted in changes in microbiologic spectrum are alterations

CHEMISTRY
Most of the available cephalosporins are semisynthetic derivatives of Cefazolin
cephalosporin C. The basic structure of the cephem nucleus includes N N S
a β-lactam ring fused to a six-member sulfur-containing dihydrothi- N CH2 C HN N N
azine ring (Fig. 21-1). The cephem nucleus is chemically distinct from N
N CH2 S CH3
the penicillin nucleus, which contains a five-member thiazolidine ring. O S
O
Basic structure numbering of the cephalosporin ring system begins
within the dihydrothiazine ring at the sulfur moiety. The starting mate- A COOH
rial used as the nucleus for current cephalosporin development is Cephalothin
7-aminocephalosporanic acid (7-ACA). Attempts to alter the physio- S O
CH2 C HN
chemical and biologic properties of the cephalosporins by chemical S
side chain modifications were based on successes with similar struc- N CH2 O C CH3
O
tural changes at the 6-aminopenicillanic acid side chain of penicillin.3 O
Chemical modifications of the basic cephem structure by substitution B COOH
of constituents at positions C1, C3, and C7 led to the various cephalo-
sporin compounds in use today.4,5 Alterations in position C7 and C3 Cefadroxil
S
are also commonly referred to as R1 and R2, respectively. In general, HO CH C HN
changes at R1 affect the microbial spectrum of activity. These modifica-
tions often have an impact on the stability of the compound to enzy- N CH3
O
NH2 O
matic destruction by β-lactamases or on its affinity for the drug target.
C COOH
Cephalexin
1 S
CH C HN
7 S
R1 C HN
N CH3
3 O
N R2 NH2 O
O
O
D COOH
COO- FIGURE 21-2  First-generation cephalosporins. A, Cefazolin. B,
FIGURE 21-1  Basic cephalosporin nucleus. Cephalothin. C, Cefadroxil. D, Cephalexin.
278
278.e1
KEYWORDS
cefaclor; cefadroxil; cefamandole; cefazolin; cefdinir; cefditoren;
cefepime; cefixime; cefmetazole; cefonicid; cefoperazone; cefotaxime;

Chapter 21  Cephalosporins


cefotetan; cefoxitin; cefpodoxime; cefpirome; cefprozil; ceftaroline;
ceftazidime; ceftibuten; ceftizoxime; ceftobiprole; ceftolozane;
ceftriaxone; cefuroxime; cefuroxime-axetil; cephalexin; cephalothin;
cephapirin; cephradine; loracarbef; moxalactam
279
Cefuroxime Cefoxitin
O H O H OCH3
S S
C C N O
CH2 C N

Chapter 21  Cephalosporins


O O
N S
N CH2 O C NH2 N
O O CH2 O C NH2
OCH3 COOH
A COOH C

O Cefotetan Cefprozil
O H OCH3 S
CH C HN CH3
H2N C S S HO
CH C N N N
N CH CH
HO C S N NH2 O
N CH2 S O
O N
O D COOH
COOH CH3
B
FIGURE 21-3  Second-generation cephalosporins. A, Cefuroxime. B, Cefotetan. C, Cefoxitin. D, Cefprozil.

Cefotaxime
O H Cefditoren
O H
S
N N O O CH3 N
C C
N CH C N
N CH2 O C CH3
N N
H2N S O N S
OCH3 H2N S O
OCH3
A COOH COOH
Ceftazidime
E
Cefixime
O H O H
S S
N C C N N CH C N
CH3 N CH2 N + N
N N
H2N S O O CH CH2
O C CH3 H 2N S
COOH OCH2COOH COOH
F
B COOH
Cefpodoxime
O H Ceftriaxone O H
S O S
N C C N N N CH C N
N CH2 S OH N
N N
S O N N H2N S O CH2 OCH3
H2N
OCH3 OCH3 COOH
COOH
CH3
G
C
Cefdinir
H2N OH Ceftibuten
S O H
N H H
H S S
N C C N N CH C N
N N N
O O H
O CH=CH2 H2N S
CO2H H OCH2COOH COOH
D
FIGURE 21-4  Third-generation cephalosporins. A, Cefotaxime. B, Ceftazidime. C, Ceftriaxone. D, Cefdinir. E, Cefditoren. F, Cefixime. G, Cefpo-
doxime. H, Ceftibuten.

at the α-carbon of the acyl side chain.5 These changes have ranged from have a similar structure at the C7 position. Ceftazidime and ceftolo-
the relatively simple addition of a hydroxyl group to the addition of zane differ from these drugs in that the methoxyimino group is
large synthetic moieties. Each of the acyl side chain alterations has led replaced with a dimethylacetic acid moiety attached to the imino
to enhanced gram-negative potency because of improved β-lactamase group.8,10 This alteration enhances activity against Pseudomonas aeru-
stability. The addition of a hydroxyl group at the α-carbon led to the ginosa but reduces activity against staphylococci. Two other modifica-
second-generation cephalosporin, cefamandole. The second-generation tions that have resulted in compounds with increased activity against
cephalosporin cefuroxime resulted from the addition of a methoxyimino P. aeruginosa are the addition of an ureido-2,3-dioxopiperazine group
group in the α position, along with a furyl ring at the β-acyl side chain. or a carboxyl group on the α-carbon, producing cefoperazone and
Addition of a 2-aminothiazol group to the C7 β-acyl side chain and a moxalactam, respectively.5 These changes are similar to those of piper-
methoxyimino group to the α-carbon led to many of the third- and acillin and carbenicillin.
fourth-generation cephalosporins (Figs. 21-4 and 21-5).8,9 Cefotaxime, Numerous modifications at R2 (the C3 position) have also played
ceftizoxime, ceftriaxone, cefepime, cefpirome, and cefpodoxime all a significant role in the development of the current cephalosporins. An
280
Cefepime Ceftaroline
O H O H
S S
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

N CH C N N CH C N S
N CH3
N N N N N
O CH2 N+ O
S
H2N S H2N S
OCH3 OCH2
COOH COOH
A CH3 A
Cefpirome Ceftobiprole
O H O H
S S
N CH C N N CH C N
N N N
N N NH
O CH2 N+ O O
H2N S OCH3 H2N S OH
B COOH B COOH

FIGURE 21-5  Fourth-generation cephalosporins. A, Cefepime. FIGURE 21-6  Methicillin-resistant Staphylococcus aureus–active
B, Cefpirome. cephalosporins. A, Ceftaroline. B, Ceftobiprole.

TABLE 21-1  Classification of Parenteral and Oral Cephalosporins


SECOND THIRD FOURTH
FIRST GENERATION GENERATION CEPHAMYCINS GENERATION GENERATION MRSA-ACTIVE
Parenteral Cephalosporins
Cefazolin (Ancef, Kefzol) Cefamandole (Mandol)* Cefmetazole (Zefazone)* Cefoperazone (Cefobid)* Cefepime (Maxipime) Ceftaroline (Teflaro)
Cephalothin (Keflin, Seffin)* Cefonicid (Monocid)* Cefotetan (Cefotan) Cefotaxime (Claforan) Cefpirome (Cefrom)* Ceftobiprole (Zeftera)*
Cephapirin (Cefadyl)* Cefuroxime (Kefurox, Cefoxitin (Mefoxin) Ceftazidime (Fortaz)
Cephradine (Velosef)* Zinacef) Ceftizoxime (Cefizox)*
Ceftriaxone (Rocephin)
Moxalactam*
Oral Cephalosporins
Cefadroxil (Duricef, Ultracef) Cefaclor (Ceclor)* Cefdinir (Omnicef)
Cephalexin (Keflex, Biocef, Cefprozil (Cefzil) Cefditoren (Spectracef)
Keftab) Cefuroxime-axetil (Ceftin) Cefixime (Suprax)
Cephradine (Velosef)* Loracarbef (Lorabid)* Cefpodoxime (Vantin)
Ceftibuten (Cedax)
*No longer marketed in the United States.

acetoxy side chain is present in cephalothin, cephapirin, and cefotax- lipophilicity at the C3 position for activity, prodrugs have been required
ime.5 Cephalosporins with this structure can be metabolized in both to enhance aqueous solubility.16,17
serum and liver to a less active desacetyl derivative. Such drugs also
tend to have a short half-life. A chloride substitution at R2 enhanced CLASSIFICATION
the gram-negative spectrum of activity and led to the development of There are several microbiologic and pharmacologic differences that
cefaclor, an early second-generation cephalosporin. The unique phar- could serve as a basis for classification among the cephalosporin drug
macology of ceftriaxone results from an R2 modification. Substitution class. The most widely accepted classification includes five divisions,
of a heterocyclic thiomethyl group at the C3 position increases biliary or generations, based loosely on the microbial spectrum of activity
secretion and remarkably prolongs the elimination half-life of the com- (Table 21-1). The first-generation cephalosporins exhibit activity
pound because of high protein binding.5,11 The addition of a positively focused primarily on gram-positive bacteria. The second-generation
charged quaternary ammonium moiety in the C3 position contributed drugs have enhanced activity against gram-negative bacilli but main-
to the development of the fourth-generation cephalosporins, cefepime tain varying degrees of activity against gram-positive cocci. The cepha-
and cefpirome.9 The chemical modification produces a zwitterion, mycin group is included in the second-generation classification. The
which enhances the ability of the compound to penetrate the outer cephamycins are noted for their additional activity against gram-
membrane of gram-negative organisms. The inclusion of additional negative anaerobic bacteria, such as Bacteroides spp.
amino groups to this ammonium moiety further enhances activity The third-generation cephalosporins have markedly increased
against P. aeruginosa.10 Not all modifications have led to desired effects. potency against gram-negative bacilli; however, for some compounds
The placement of a thiomethyl tetrazole ring (MTT) at the R2 position in this group, activity against gram-positive cocci is reduced. Among
enhanced antibacterial activity but also resulted in two important the third-generation group, a few compounds, such as ceftazidime and
adverse effects that have limited use of these compounds.12,13 Cefaman- ceftolozane, are considered separately for activity against P. aeruginosa.
dole, cefotetan, cefoperazone, and moxifloxacin (an oxycephem) The fourth generation has the widest spectrum of activity of the five
contain this MTT side chain, which is responsible for coagulation groups. These drugs, such as cefepime and cefpirome, have activity
abnormalities related to antagonism of vitamin K action. This side against most gram-negative bacilli, including P. aeruginosa, and main-
chain is also responsible for the disulfiram-like properties of these tain their potency against gram-positive cocci. The third- and fourth-
compounds. generation drugs combined are also called the extended-spectrum
More recently, cephalosporins with enhanced activity against cephalosporins.
methicillin-resistant Staphylococcus aureus (MRSA) have been devel- The fifth group is referred to as the MRSA-active cephalosporins
oped (Fig. 21-6). A variety of structural alterations at the C3 and and currently includes ceftaroline and ceftobiprole (not yet available in
C7 positions have increased the drug’s stability to β-lactamase inactiva- the United States). In addition to this unique activity against MRSA
tion and enhanced tight binding to the altered penicillin-binding among the cephalosporins, these drugs also have enhanced activity
protein 2a′.14,15 Because some of these compounds require more against Streptococcus pneumoniae and Enterococcus faecalis. Their
281
activity against gram-negative bacilli is similar to that of the third- The first-generation cephalosporins are not very active against Hae-
generation cephalosporins. Ceftobiprole also has activity against P. mophilus influenzae or Moraxella catarrhalis. The second-generation
aeruginosa. drugs are about fourfold more potent against these respiratory patho-
More recently, a few cephalosporins susceptible to inactivation by gens. The third-generation cephalosporins have the lowest MICs for

Chapter 21  Cephalosporins


extended-spectrum β-lactamases (ESBLs) have been combined with H. influenzae and M. catarrhalis—10 to 100 times lower than those
β-lactamase inhibitors to expand their activity against gram-negative of the second-generation drugs. The first-generation cephalosporins
bacilli. Ceftazidime and ceftaroline are combined with avibactam, also are not as active against Neisseria spp. as the second-, third-, and
whereas ceftolozane is combined with tazobactam.18 fourth-generation drugs.
Although all of the cephalosporins are considered to be active
MECHANISM OF ACTION against Escherichia coli, Klebsiella pneumoniae, and Proteus mirabilis,
The mechanism of antibacterial activity of cephalosporins is similar to the potency of the third- and fourth-generation drugs and the MRSA-
that of other β-lactam drugs. Bacterial growth is inhibited by interfer- active cephalosporins is 10- to 100-fold greater than that of the first-
ence with the synthesis of the cell wall. The primary target of these and second-generation cephalosporins. The increased potency of the
compounds within the cell wall is the peptidoglycan cross-linkage later-generation drugs extends to strains of Enterobacter, Serratia,
structure.19 Peptidoglycans are polysaccharide chains consisting of Citrobacter, and Morganella spp., which are usually resistant to the
alternating N-acetylglucosamine and N-acetylmuramic acid residues. first- and second-generation drugs. Several cephalosporins, such as
The polysaccharide chains are cross-linked at the pentapeptide side ceftazidime, ceftolozane, cefoperazone, cefepime, cefpirome, and cef-
chain of the N-acetylmuramic acid residues to form a netlike structure. tobiprole, are active against many strains of P. aeruginosa. The third-
These structures are inserted into the cytoplasmic membrane from the and fourth-generation cephalosporins also exhibit enhanced potency
cytoplasm by the action of a group of enzymes that includes transpep- against strains of Salmonella and Shigella.
tidases, carboxypeptidases, and endopeptidases. The lactam ring pro- Many cephalosporins are active against penicillin-susceptible
vides for penicillins and cephalosporins a conformation similar to the gram-positive anaerobes, such as peptostreptococci.64,65 Among the
terminal d-alanine-d-alanine of the pentapeptide.20 The antibiotics cephalosporins, the best activity against Bacteroides fragilis is with
bind covalently to these enzymes, in particular to the transpeptidases, drugs such as the cephamycins—cefotaxime, ceftriaxone, and ceftizox-
resulting in loss of enzyme activity.19 The enzyme drug targets are ime. Many of the drugs are active against spirochetes, including the
referred to as penicillin-binding proteins (PBPs).21,22 agents of Lyme disease and syphilis.66,67As a group, the cephalosporins
The location of the PBPs relative to the extracellular space differs have very poor activity against Chlamydia, Mycoplasma, and Listeria
between gram-positive and gram-negative bacteria. The peptidoglycan spp.68-70
of gram-positive bacteria is located on the outer surface of the cell. A
complex lipopolysaccharide (LPS) structure is located on the outer- MECHANISMS OF RESISTANCE
most surface of gram-negative bacteria; cephalosporins must first pen- Four general mechanisms can result in resistance to cephalosporin
etrate or diffuse across the LPS membrane to reach the PBPs. The PBP antibiotics: (1) antibiotic destruction by hydrolyzing β-lactamase
targets within bacteria vary by type and amount. These targets are enzymes, (2) reduced penetration of the antibiotic through the LPS
numbered by convention on the basis of molecular weight, with letters membrane to the PBP target, (3) enhanced efflux of the drug from the
differentiating proteins of similar molecular weight. Gram-positive periplasmic space, and (4) alteration in the PBP target, resulting in
and gram-negative cocci typically have 3 to 5 PBPs; gram-negative reduced binding affinity. Most often, resistance in a bacterial popula-
bacilli usually contain 7 to 10 PBPs. The cephalosporin drugs can vary tion is caused by a single mechanism; however, an increasing percent-
in their affinity for each of these drug targets. At low concentrations, age of organisms are exhibiting multiple mechanisms.71
cephalosporins preferentially bind to PBP 3 in gram-negative bacilli, Production of β-lactamase enzymes that hydrolyze the β-lactam
resulting in filament formation with septae.15,23 The events after the ring is a predominant resistance mechanism for many gram-negative
covalent binding of cephalosporins to the PBP targets that lead to cell bacteria. With staphylococci, most cephalosporins, with the exception
lysis and death are not entirely understood. of cephaloridine, are poorly hydrolyzed by staphylococcal penicillin-
In general, cephalosporins are considered to be bactericidal drugs. ases. Resistance to cephalosporins in these organisms is due almost
The rate of killing of bacteria by cephalosporins exhibits minimal entirely to reduced binding affinity of the PBPs. Although all gram-
dependence on the concentration of the antibiotic.24 Maximal bac­ negative bacilli produce β-lactamase enzymes, the type and amount of
terial killing is observed at concentrations four times the minimal enzyme varies among organisms. These enzymes are located in the
inhibitory concentration (MIC). Cephalosporins produce persistent periplasmic space between the outer LPS membrane and the inner cell
suppression of bacterial growth (i.e., the postantibiotic effect) of membrane. Drugs that are able to penetrate the outer membrane can
several hours’ duration with gram-positive bacteria, but they induce be degraded before reaching the PBP target. The net antimicrobial
very short or no postantibiotic effects with gram-negative bacilli.25,26 activity of cephalosporins against gram-negative bacilli is dependent
The duration of time during which drug concentrations exceed the on the rate of penetration across the outer membrane and the stability
MIC is the major determinant of the antibacterial activity of the of the drug to the various hydrolyzing β-lactamases. The penetration
cephalosporins.26-28 of drugs across the outer membrane is through water-filled channels
formed by various membrane proteins, termed porins. Movement
SPECTRUM OF ACTIVITY through porin channels depends on the size, shape, charge, and hydro-
The cephalosporins are active against a wide variety of aerobic and philic properties of the compound. The concentration of a drug with
anaerobic bacteria (Tables 21-2 and 21-3).29-63 Most drugs are active a slow rate of penetration is low relative to the amount of β-lactamase
against streptococci and staphylococci. Some of the differences in within the periplasmic space, so the relatively high β-lactamase con-
potency among the agents are magnified by their activity against centration can enhance antibiotic inactivation. For a drug that can
penicillin-resistant pneumococci. Ceftaroline and ceftobiprole have penetrate rapidly, the converse is true. For example, the zwitterion of
the greatest potency against this organism, followed by cefditoren, cefepime enhances movement across the membrane, resulting in high
ceftriaxone, cefotaxime, cefepime, and cefpirome.59,63 The cephamy- concentrations in the periplasmic space and a relative net resistance to
cins, ceftazidime, cefixime, and ceftibuten have the poorest activity drug hydrolysis.72
against methicillin-susceptible staphylococci. Although MRSAs are The number of unique β-lactamases is markedly increasing with
resistant to all the earlier cephalosporins, the new MRSA-active cepha- more than 1000 distinct enzymes. Some of this increase is due to point
losporins, ceftaroline and ceftobiprole, exhibit MICs of approximately mutations in the β-lactamases. These proteases may be genetically
0.5 to 4 µµg/mL with such strains.59,63 Enterococci have also consis- encoded chromosomally or extrachromosomally. Stable derepression
tently been resistant to the cephalosporins, with most MICs greater of a chromosomal mutation is a common genotypic scenario. This is
than 32 µg/mL. However, the new MRSA-active cephalosporins have observed predominantly in Enterobacter spp., Serratia spp., Citrobacter
much lower MICs for ampicillin-susceptible strains. These have ranged freundii, and P. aeruginosa.73 Transmissible plasmids have acquired the
from 0.12 to 4 µg/mL for both drugs.59,63 genes for AmpC enzymes and are appearing in bacteria, such as E. coli
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

TABLE 21-2  In Vitro Activity of Cephalosporins against Selected Gram-Positive Cocci, Haemophilus influenzae, Moraxella catarrhalis, and Neisseria
Species*
STREPTOCOCCUS VIRIDANS STAPHYLOCOCCUS
PNEUMONIAE STREPTOCOCCUS STREPTOCOCCUS STREPTOCOCCI AUREUS STAPHYLOCOCCUS HAEMOPHILUS MORAXELLA NEISSERIA NEISSERIA
CEPHALOSPORIN (PSSP) (PRSP) PYOGENES AGALACTIAE GROUP (MSSA) (MRSA) EPIDERMIDIS INFLUENZAE CATARRHALIS MENINGITIDIS GONORRHOEAE
First Generation
Cefazolin 0.5/4 32/>32 0.12/0.12 0.12/0.12 0.12/0.12 0.5/2 R 0.5/>32 4/16 2/4 — 16/32
Cephalothin 0.12/0.25 8/16 0.05/0.10 0.12/0.5 0.25/0.50 0.12/0.5 R 0.5/32 4/8 4/8 0.25/0.5 8/32
Cefadroxil (O) 2/4 >32 0.12/0.25 0.25/2 — 2/8 R 4/>32 16/>32 2/4 — 8/64
Cephalexin (O) 1/2 >32 0.25/2 0.5/4 — 2/4 R 1/>32 8/16 2/8 2/2 4/16
Cephradine (O) 2/4 >32 0.25/2 0.5/2 — 1/4 R 4/>32 4/16 2/4 — 8/16
Second Generation
Cefamandole 0.12/0.5 8/>32 0.12/0.12 0.12/0.5 0.12/4 1/1 R 0.5/>32 2/8 1/4 0.12/0.12 0.25/4
Cefonicid 0.5/1 4/>32 0.12/0.12 0.12/2 0.12/8 1/2 R 2/>32 0.5/1 1/4 0.12/0.12 0.06/0.5
Cefuroxime 0.12/0.25 4/>32 0.12/0.12 0.12/0.12 0.12/0.5 1/2 R 0.5/>32 1/2 0.5/2 0.12/0.12 0.015/0.25
Cefaclor (O) 0.5/1 16/>32 0.06/0.5 0.5/2 — 1/8 R 1/>32 2/32 0.5/2 0.06/0.25 0.25/16
Cefprozil (O) 0.12/0.5 8/>32 0.03/0.12 0.06/0.25 — 0.5/2 R 0.25/32 2/16 1/8 — 0.12/4
Loracarbef (O) 0.5/2 >32 0.5/2 0.5/2 — 1/4 R 4/>32 1/4 0.5/4 0.12/0.25 0.5/4
Cephamycins
Cefmetazole 2/16 >32 0.5/0.5 2/2 2/4 4/16 R 8/>32 1/4 0.12/0.5 0.12/0.12 0.25/4
Cefotetan 8/16 >32 2/4 4/8 2/8 8/16 R 32/>32 1/2 0.12/2 0.12/0.25 0.25/2
Cefoxitin 2/4 32/>32 1/2 2/2 4/16 4/8 R 2/>32 1/4 0.25/0.5 0.12/0.25 0.25/4
Third Generation
Cefoperazone 0.06/0.12 4/16 0.12/0.12 0.12/0.25 0.5/1 2/4 R 2/>32 0.015/0.25 0.12/2 0.12/0.5 0.03/0.06
Cefotaxime 0.015/0.06 0.5/2 0.015/0.015 0.03/0.25 0.06/0.25 2/2 R 4/>32 0.008/0.015 0.5/1 0.004/0.008 0.004/0.008
Ceftazidime 0. 25/1 16/>32 0.12/0.25 0.25/0.5 1/2 8/32 R 8/>32 0.06/0.12 0.03/0.5 0.015/0.06 0.03/0.06
Ceftizoxime 0.25/1 16/32 0.015/0.015 0.12/0.12 0.25/2 4/8 R 4/>32 0.015/0.03 0.03/0.5 0.008/0.03 0.008/0.015
Ceftriaxone 0.03/0.06 10.5/2 0.015/0.03 0.03/0.06 0.06/0.25 2/4 R 4/>32 0.008/0.015 0.25/0.5 0.008/0.015 0.002/0.004
Moxalactam 1/1 — 1/2 — — 8/16 R 8/>32 0.03/0.12 0.03/0.12 0.008/0.06 0.015/0.06
Cefdinir (O) 0.06/0.12 2/8 0.015/0.03 0.03/0.06 — 0.25/0.5 R 0.25/>32 0.12/0.5 0.06/0.25 0.06/0.25 0.008/0.06
Cefditoren (O) 0.015/0.03 0.5/2 0.008/0.015 0.06/1 — 0.25/1 R 0.25/>32 0.008/0.015 0.25/1 <0.06/ 0.06 0.004/0.06
Cefixime (O) 0.25/1 32/>32 0.06/0.25 0.12/0.25 — 16/>32 R 16/>32 0.015/0.12 0.03/0.5 <0.06/0.06 0.015/0.06
Cefpodoxime (O) 0.015/0.06 2/>32 0.06/0.12 0.03/0.12 — 2/4 R 2/>32 0.015/0.12 1/2 <0.06/0.06 0.06/0.06
Ceftibuten (O) 4/8 >32 0.5/2 4/16 — 16/>32 R 16/>32 0.06/0.12 2/4 <0.06/0.25 0.015/0.5
Fourth Generation
Cefepime 0.06/0.12 0.5/2 0.015/0.12 0.05/0.05 0.016/0.03 2/4 R 2/>32 0.06/0.12 1/4 0.03/0.06 0.03/0.06
Cefpirome 0.03/0.12 0.5/2 0.008/0.06 0.06/0.06 0.06/0.25 1/2 R 1/>32 0.06/0.12 0.5/2 0.06/0.06 .015/0.12
MRSA-Active
Ceftaroline 0.008/0.015 0.12/0.25 0.008/0.015 0.015/0.03 0.008/0.03 0.25/0.25 0.5/1 0.5/0.5 1/4 0.008/0.03 0.002/0.004 0.008/0.03
Ceftobiprole 0.008/0.015 0.25/0.25 0.008/0.015 0.06/0.12 0.06/0.12 0.25/0.5 0.5/2 0.5/1 0.5/4 0.06/0.25 0.002/0.004 0.03/0.06
*Minimal inhibitory concentration (MIC) for 50% and 90% of strains in µg/mL.
MRSA, methicillin-resistant Staphylococcus aureus; MSSA, methicillin-susceptible S. aureus; (O), oral; PRSP, penicillin-resistant Streptococcus pneumoniae; PSSP, penicillin-susceptible S. pneumoniae; R, resistant (MICs are variable, but organism is resistant in patients).
TABLE 21-3  In Vitro Activity of Cephalosporins against Selected Aerobic and Anaerobic Gram-Negative Bacilli*
ESCHERICHIA KLEBSIELLA PROTEUS ENTEROBACTER ENTEROBACTER CITROBACTER SERRATIA PSEUDOMONAS MORGANELLA BACTEROIDES SALMONELLA SHIGELLA
CEPHALOSPORIN COLI PNEUMONIAE MIRABILIS AEROGENES CLOACAE FREUNDII SP. AERUGINOSA SP. FRAGILIS SP. SP.
First Generation
Cefazolin 2/16 2/>16 4/16 >32 >32 >32 >32 >32 >32 >32 2/4 2/8
Cephalothin 4/8 1/16 8/16 >32 >32 >32 >32 >32 >32 >32 2/4 4/8
Cefadroxil (O) 4/>16 8/>16 16/>32 >32 >32 >32 >32 >32 >32 >32 8/>16 4/16
Cephalexin (O) 8/>16 8/32 16/>32 >32 >32 >32 >32 >32 >32 >32 4/16 8/>16
Cephradine (O) 4/>16 4/>16 16/>32 >32 >32 >32 >32 >32 >32 >32 4/>16 8/>16
Second Generation
Cefamandole 1/2 1/8 1/2 4/>32 2/>32 2/>32 16/>32 >32 4/>32 32/>32 0.5/4 0.5/2
Cefonicid 2/8 2/8 1/2 4/>32 8/>32 8/>32 >32 >32 16/>32 32/>32 2/8 2/8
Cefuroxime 2/8 2/16 2/4 8/>32 8/>32 8/>32 >32 >32 32/>32 8/>32 4/8 2/4
Cefaclor (O) 2/>16 2/32 2/4 >32 >32 >32 >32 >32 >32 >32 2/8 4/16
Cefprozil (O) 2/8 1/>32 2/2 >32 >32 16/>32 16/>32 >32 16/>32 >32 2/8 4/16
Loracarbef (O) 1/>16 0.5/8 0.5/2 16/>32 16/>32 4/>32 >32 >32 32/>32 >32 0.5/8 0.25/8
Cephamycins
Cefmetazole 0.5/1 1/2 2/4 >32 >32 >32 16/>32 >32 4/8 8/>32 0.5/2 1/2
Cefotetan 0.12/0.5 0.12/0.5 0.12/0.5 32/>32 8/>32 0.5/>32 1/8 >32 2/4 8/>32 0.12/0.12 0.12/0.5
Cefoxitin 2/8 2/8 2/4 >32 >32 >32 16/>32 >32 8/16 8/32 2/4 2/4
Third Generation
Cefoperazone 0.12/8 0.25/8 0.5/1 0.25/8 0.25/8 0.5/32 2/8 4/>32 1/8 32/>32 0.5/4 0.25/1
Cefotaxime 0.06/0.25 0.06/0.25 0.06/0.25 0.12/16 0.25/32 0.25/>32 0.25/2 16/>32 0.25/4 8/>32 0.06/0.12 0.06/0.25
Ceftazidime 0.06/0.25 0.25/1 0.06/0.5 0.25/32 0.25/>32 0.5/>32 0.25/2 2/16 0.12/0.5 >32 0.12/0.5 0.06/0.25
Ceftizoxime 0.03/0.12 0.03/0.12 0.008/0.015 0.06/16 0.06/16 0.25/>32 0.12/0.5 32/>32 0.25/2 16/>32 0.015/0.25 0.008/0.25
Ceftriaxone 0.06/0.12 0.06/0.25 0.008/0.03 0.25/16 0.25/>32 0.12/>32 0.25/4 32/>32 0.008/0.25 8/>32 0.06/0.25 0.03/0.12
Moxalactam 0.12/0.25 0.12/0.25 0.25/0.5 0.25/16 0.5/8 0.25/8 0.25/4 32/>32 0.25/0.5 2/32 0.12/0.25 0.12/0.25
Cefdinir (O) 0.12/0.25 0.06/0.25 0.06/0.12 0.5/>32 0.5/>32 0.25/>32 4/32 16/>32 4/16 16/>32 0.12/0.25 0.25/0.5
Cefditoren (O) 0.25/0.5 0.25/1 0.12/1 0.5 />32 1/>32 1/>32 2/32 >32 — 4/>32 0.25/0.5 0.25/0.5
Cefixime (O) 0.12/0.25 0.03/0.12 0.008/0.03 0.5/>32 0.12/>32 2/>32 2/>32 >32 2/32 16/>32 0.06/0.25 0.25/0.5
Cefpodoxime (O) 0.25/1 0.5/2 0.06/0.12 2/>32 4/>32 2/>32 1/8 >32 2/>32 16/>32 0.5/1 0.12/0.25
Ceftibuten (O) 0.12/0.25 0.06/0.25 0.015/0.03 1/32 2/>32 1/>32 0.5/8 >32 0.25/8 16/>32 0.06/0.25 0.06/0.25
Fourth Generation
Cefepime 0.03/0.06 0.03/0.25 0.06/0.12 0.06/0.5 0.06/2 0.06/2 0.12/0.5 2/16 0.03/0.12 >32 0.06/0.12 0.03/0.06
Cefpirome 0.06/0.12 0.06/0.25 0.06/0.12 0.06/0.5 0.12/4 0.03/2 0.25/2 2/16 0.03/0.12 32/>32 0.06/0.25 0.06/0.12
MRSA-Active
Ceftaroline 0.06/1 0.06/0.25 0.06/4 0.12/1 0.12/0.25 0.5/8 0.06/16 16/>32 4/>16 16/>32 0.12/>32 0.03/0.12
Ceftobiprole 0.06/0.12 0.06/0.5 0.03/0.12 0.06/1 0.03/0.5 0.12/4 0.06/ 0.12 2/16 4/>32 8/>32 0.03/0.06 0.03/0.06
*Minimal inhibitory concentration (MIC) for 50% and 90% of strains in µg/mL.
MRSA, methicillin-resistant Staphylococcus aureus; (O), oral.

Chapter 21  Cephalosporins


284
and K. pneumoniae, that lack or poorly express a chromosomal Changes in the PBP target are responsible for reduced cephalospo-
enzyme.74 The AmpC cephalosporinase is capable of inactivating rin affinity and subsequent drug resistance for S. pneumoniae, MRSA,
almost all current cephalosporins, including the cephamycins. Emer- H. influenzae, and some Neisseria gonorrhoeae strains. Changes in the
gence of this type of resistance is frequent when infections resulting PBP target may result from amino-acid substitutions or insertions.
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

from these organisms are treated only with broad-spectrum Almost 40 amino-acid substitutions in PBP 2b have been described in
cephalosporins.75,76 the pneumococcus.103,104 Multiple mutations have also been described
Many of the newer enzymes represent variants of the common in PBP 2a′ of a strain of MRSA resulting in resistance to ceftobiprole.105
plasmid-encoded Temoneira (TEM)-1, TEM-2 and sulfhydryl variable Resistance in S. pneumoniae and several other pathogens has also
(SHV)-1 β-lactamases. These enzymes have been observed most com- resulted from the insertion of resistance sequences from other related
monly in K. pneumoniae and E. coli and are referred to as extended- species.
spectrum β-lactamases (ESBLs) because they are capable of inactivating
many third- and fourth-generation cephalosporins.77,78 The new PHARMACOLOGIC PROPERTIES
MRSA-active cephalosporins are also susceptible to inactivation by The pharmacologic properties of selected cephalosporins are listed
ESBLs.79,80 These enzymes are the result of amino-acid substitutions in Table 21-4.106-110,111-132,133-135 Cephalosporins are polar, water-soluble
related to point mutations in the common β-lactamase genes (i.e., compounds. Within each of the first-, second-, and third-generation
TEM and SHV). SHV ESBLs are the most common ESBLs found in K. classifications, there are both oral and parenteral formulations. The
pneumoniae.81 More recently, the cefotaxime/ceftazidime-hydrolyzing fourth-generation compounds and those with activity against MRSA
(CTX-M) family of β-lactamases, derived from the chromosomal are available for parenteral use only. The parenteral formulations are
enzyme of Kluyvera spp., have spread into K. pneumoniae and E. coli.82 available for both intravenous and intramuscular administration. All
This worldwide spread of these organisms is complicating initial treat- of the parenteral formulations, with the exception of cephradine, are
ment of urinary tract infections in many regions.83 Gram-negative stable in solution at room temperature for 24 hours or longer.136,137
bacilli producing carbapenemases are also emerging in certain parts of Drug stability at room temperature facilitates use of these compounds
the world. These enzymes, often called K. pneumoniae carbapenemases for home intravenous therapy, including continuous infusions. Many
(KPCs), are capable of hydrolyzing most of the cephalosporins.84 of the parenteral compounds can also be administered by the intra-
The different β-lactamase enzymes can vary significantly in their peritoneal route for treatment of peritoneal infections associated with
affinity for drugs within the cephalosporin class. For example, cefepime, continuous ambulatory peritoneal dialysis.138 Formulations of the oral
cefpirome, and ceftobiprole are less susceptible than other cephalospo- cephalosporins are available as tablets, capsules, or suspensions.
rins to inactivation by AmpC cephalosporinases.79,85,86 The cephamy- In contrast to many other β-lactams, oral preparations of the cepha-
cins are not susceptible to inactivation by ESBLs. Ceftazidime and losporins are stable in the acid milieu of the upper gastrointestinal
cefpodoxime generally exhibit high sensitivity to inactivation by tract. Cephalosporins can be actively absorbed if their structure medi-
ESBLs.77 On the other hand, cefepime, cefpirome, and even cefotaxime ates transport by the dipeptide and tripeptide transport systems in the
and ceftriaxone may exhibit only modest inactivation, and the organ- brush border membrane of the small intestine.139 Cephalexin, cephra-
ism may still be considered susceptible by standard susceptibility dine, cefadroxil, cefaclor, cefprozil, and loracarbef have an aminoben-
testing. zyl group or a similar group in the C7 position and have high oral
Tests using ceftazidime, cefpodoxime, cefotaxime, or ceftriaxone to bioavailability (80% to 95%). Ceftibuten, cefixime, and cefdinir have
identify strains of K. pneumoniae, E. coli, and P. mirabilis producing other groups in the C7 position and exhibit more variable bioavail-
ESBLs have been devised for clinical laboratories by the Clinical and ability after oral dosing. Absorption by the dipeptide and tripeptide
Laboratory Standards Institute (CLSI).87 However, the identification of transport systems appears to be both pH and calcium dependent.139
ESBLs in the clinical laboratory is complicated by new emerging ESBLs Drugs that have low oral bioavailability can be esterified to enhance
called the CTX-M enzymes, which hydrolyze ceftazidime much less absorption; the ester prodrug is hydrolyzed after absorption in the
than other third- and fourth-generation cephalosporins.88-90 The previ- intestinal epithelial cells. The esters commonly used include axetil,
ous recommendation of the CLSI was that all such strains should be proxetil, and pivoxil formulations.140 Ester prodrug formulations exist
called resistant to cephalosporins other than the cephamycins, even if for cefuroxime, cefditoren, and cefpodoxime. Absorption of the ester
they are susceptible on standard susceptibility tests.87 This recommen- is still not complete; in fact, the percent oral bioavailability of ester
dation was in line with a large, prospective observational study of formulations is lower than that of most nonesterified compounds.
patients with Klebsiella bacteremia that reported a significantly poorer Absorption of the ester formulations is enhanced by concomitant food
outcome in patients treated with third- and fourth-generation cepha- intake140 because food within the stomach delays gastric emptying and
losporins, compared with carbapenems.91 The poorer outcome was prolongs contact with the mucosal surface.
observed primarily in strains with MICs greater than 2 µg/mL. The Distribution of cephalosporins within the body is governed by
current susceptibility breakpoints for cephalosporins have been the lipid solubility of the drug and the extent of protein binding.
lowered in the United States, as has already been accomplished in β-Lactams bind almost exclusively to albumin. The extent of protein
Europe.92,93 According to CLSI, strains with ESBLs that have susceptible binding can vary from less than 10% to as much as 98%.126 Because
MICs are considered treatable with cephalosporins at the cited dosages, only unbound drug can pass through capillary pores into interstitial
but there are some recent studies that would support lower MIC sus- fluid or across cell membranes into intracellular fluid, avidly bound
ceptibility breakpoints for cefepime. 94,95 compounds tend to exhibit high serum concentrations and low tissue
It is unlikely that deletion or mutation of porin proteins causes concentrations. In general, the cephalosporins are largely confined to
primary resistance to cephalosporins. However, such changes can alter the extracellular compartment. Drug concentrations in subcutaneous
the relationship between the concentrations of drug and β-lactamase blisters, a model for extracellular drug penetration, are similar to those
in the periplasmic space, resulting in much more hydrolysis of the found in serum.126,141 Techniques measuring extracellular drug con­
cephalosporin. For example, strains of K. pneumoniae containing centrations in human tissues (e.g., microdialysis) have demonstrated
ESBLs have been shown to be resistant to cephamycins because of the that concentrations of unbound drug in interstitial fluid are also similar
lack of an outer membrane porin protein.96 Porin-deficient strains are to those in serum.142,143 The cephalosporins have relatively poor intra-
especially high in Enterobacter aerogenes.97 cellular concentrations. Tissue homogenates, which mix intracellular
The endogenous AcrAB multidrug efflux system in E. coli affects and extracellular fluid, always provide concentrations that are lower
the potency of penicillins but has little effect on the activity of cepha- than those in serum because of dilution by the larger intracellular
losporins.98 However, resistance in P. aeruginosa has been associated volume.141 This group of compounds does not achieve intracellular
with the MexAB-OprM efflux pump for ceftazidime and the MexXY- concentrations adequate to treat most intracellular pathogens (e.g.,
Opr efflux pump for cefepime and ceftobiprole.99-101 The differences Legionella spp.).144
among these organisms in the impact of somewhat similar pumps are In the absence of infection, drug concentrations in the cerebro­
probably due to markedly higher outer membrane permeability in E. spinal fluid (CSF) and in the vitreous humor are low. None of the
coli compared with P. aeruginosa.102 oral cephalosporins achieve therapeutic concentrations in the CSF.
TABLE 21-4  Pharmacokinetics of Cephalosporins
PEAK SERUM SERUM CSF CSF STABILITY AT ROOM
CONCENTRATION HALF-LIFE PROTEIN ROUTE OF CONCENTRATION PENETRATION TEMPERATURE (hr) or
CEPHALOSPORIN ADULT DOSE (µg/mL) (hr) BINDING (%) EXCRETION RANGE (µg/mL) (%) ORAL BIOAVAILABILTY (%)
First Generation
Cefazolin 0.5-1 g q8h 193 (1 g) 1.9 74-86 R (65%-100%) <0.7 0-4 24
Cephalothin 0.5-2 g q4-6h 64 (1 g) 0.5-1.0 50-80 R (50%-70%) 0.16-0.31 1 24
Cephapirin 0.5-2 g q4-6h 70 (1 g) 0.6 50-60 R (60%-85%) NA NA 24
Cephradine 0.5-1 g q6h 50 (1 g) 0.7 8-17 R (75%-100%) NA NA 2-10
Second Generation
Cefamandole 0.5-2 g q6h 88 (1 g) 0.7-1.3 50-78 R (80%) 0.35-7.4 0-8.6 24
Cefonicid 0.5-1 g q24h 221 (1 g) 4.4 98 R (95%) NA NA 24
Cefuroxime 0.75-1.5 g q8h 39 (1 g) 1.2-1.8 33-50 R (70%-100%) 0.35-22.5 11.6-13.7 24
Cephamycins
Cefmetazole 2 g q8h 143 (2 g) 1.3-1.8 68 R (75%-85%) NA NA 24
Cefotetan 1-2 g q12h 158 (2 g) 3.5 76-90 R (80%) 1.1-4.8 0.8-3.6 24
Cefoxitin 1-2 g q6h 110 (1 g) 0.8-1 41-79 R (90%) 1.2-22 0.8-35 24
Third Generation
Cefoperazone 1-3 g q8-12h 153 (1 g) 1.6-2.1 90 H (80%) <0.8-119 2.5-5.9 12
R (20%)
Cefotaxime 1-2 g q12h 102 (1 g) 1-1.2 35-40 R (50%-80%) 1-83 4-55 24
Ceftazidime 1-2 g q8-12h 107 (1 g) 1.5-2 17 R (80%-90%) 1.4-30 14-45 24
Ceftizoxime 1-2 g q6-12h 113 (1 g) 1.4-1.7 31 R (70%-100%) <0.5-29 3-22.6 24
Ceftriaxone 1-2 g q12-24h 145 (1 g) 6.4 85-95 R (50%) 2-20 1.5-7 72
H (40%)
Moxalactam 1-2 g q8h 70 (1 g) 2.2 50 R (67%-88%) 0.8-39 12-69 24
Ceftolozane 1-1.5 g q8-12h 90 (1 g) 2.2-2.7 <20 R (85%-93%) NA NA 24
Fourth Generation
Cefepime 1-2 g q8-12h 79 (1 g) 2 16-19 R (85%) 5.7 11.8 24
Cefpirome 1-2 g q8-12h 80 (1 g) 2 10 R (90%) 0.8-4.2 5-67 24
MRSA-Active
Ceftaroline 0.6 q q8-12h 21 (0.6 g) 2.6 7-19 R (85%) NA NA 24
Ceftobiprole 0.5 g q8-12h 34 (0.5 g) 2.85 17 R (82%) NA NA 24
Oral: First Generation
Cefadroxil 0.5-1g q12h 15 (0.5 g) 1.3-1.6 20 R (90%) NA NA 80%
Cephalexin 0.5-1g q6h 5.8 (250 mg) 0.5-1.2 6-15 R (80%-100%) NA NA 90%
Cephradine 0.5-1g q6h 15 (0.5 g) 1-2 10-20 R (80%-90%) NA NA 95%
Oral: Second Generation
Cefaclor 250-500 mg q8h 6 (250 mg) 0.5-1 25-50 R (50%-80%) NA NA 50%-90% FE
375 mg q12h 19.2 (400 mg)
Cefprozil 500 mg q12h 9.3 (500 mg) 1.3 35-45 R (61%) NA NA 95%
Cefuroxime (axetil) 250-500 mg q12h 4.6 (250 mg) 1.2 33-50 R (66%-100%) NA NA 52%-68% FE
Loracarbef 200-400 mg q12h 8 (200 mg) 1.0 25 R (87%) NA NA 90%
Oral: Third Generation
Cefdinir 300 mg q12h 2.9 1.5-1.7 60-73 R (18%) NA NA 25%
Cefditoren (pivoxil) 200-400 mg q12h 2.5 (200 mg) 0.8-1.6 88 R (16%-22%) NA NA 17% FE
Cefixime 200-400 mg 2.8 (200 mg) 3-4 65-70 R (50%) NA NA 40%-50%
q12-24h 4.5 (400 mg) H (5%)
Cefpodoxime (proxetil) 200 mg q12h 2.2 (200 mg) 2.2-2.7 18-40 R (29%-33%) NA NA 50%-80% FE
Ceftibuten 400 mg q24h 15 (400 mg) 2.4 65-77 R (57%) NA NA 75%-90%
CSF, cerebrospinal fluid; FE, food effect; H, hepatic; MRSA, methicillin-resistant Staphylococcus aureus; NA, not applicable; R, renal.
286
Penetration of most parenteral drugs from the first- and second- Most cephalosporins are eliminated by the kidney, with half-lives
generation groups is similarly poor. Parenteral cefuroxime is an excep- of 1 to 2 hours. The major mechanism for renal excretion of many
tion, and this drug also has the lowest MICs for common meningeal compounds is tubular secretion. This active transport process is largely
pathogens among the first- and second-generation cephalosporins.129 unaffected by protein binding.129 Probenecid inhibits this organic acid
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

The parenteral third- and fourth-generation drugs, such as ceftriaxone, transport system and can prolong the half-life of these compounds. For
cefotaxime, ceftazidime, and cefepime, achieve concentrations that several compounds, glomerular filtration is more important, and
would allow treatment of central nervous system infections.130-133,134,135 protein binding can significantly prolong their elimination half-life.149
The presence of an active transport system that transports many cepha- For some drugs, the elimination half-life is 3 to 8 hours, allowing for
losporins from the CSF back to serum contributes to the low drug 12- and 24-hour dosing intervals. A few compounds with high protein
levels in the CSF observed with many of the earlier-generation drugs. binding and high molecular weights, such as ceftriaxone and cefopera-
The transport protein involved in this system is similar to the protein zone, are eliminated to a large extent by the biliary route.150 Between
involved in renal secretion of β-lactam antibiotics.145 Ceftriaxone, cefo- 50% and 70% of the active parent compound may be recovered in the
taxime, ceftazidime, and cefepime exhibit minimal renal tubular secre- bile and eventually in the feces.
tion and are poor substrates for the choroid plexus pump, contributing The maximal daily doses of agents eliminated primarily by the
to higher CSF concentrations. Probenecid is a competitive substrate kidney must be reduced for patients with renal impairment. Most
for this pump and can produce higher concentrations with drugs that often, this adjustment includes both a reduction in dose level and
are effluxed by this transport system.146 Infection results in higher CSF lengthening of the dosing interval. Recommended dose adjustments
levels because inflammation can enhance penetration as well as inter- for various degrees of renal impairment and for patients receiving
fere with efflux by active transport. dialysis are presented in Table 21-5 for the currently available cepha-
Very few drugs from the cephalosporin class are extensively metab- losporins.151,152 For drugs eliminated by the biliary system, such as
olized. The three exceptions are cefotaxime, cephalothin, and cephapi- ceftriaxone, dose adjustments are unnecessary unless concomitant
rin, which undergo deacetylation of the acetoxymethyl side chain in severe hepatic insufficiency and renal insufficiency are present.151 Most
the liver.147,148 The metabolic desacetyl products still possess modest of the cephalosporins excreted by the renal route are eliminated by
microbiologic activity. The elimination half-life of desacetylcefotaxime hemodialysis. Between 20% and 50% of the parent compound is
is significantly longer than that of the parent compound, allowing less removed after a usual dialysis session. It is recommended that the
frequent administration of this cephalosporin.148 The remaining drugs drug be given again after hemodialysis. On the other hand, few cepha-
in the cephalosporin class are excreted from the body unchanged. losporins are removed to any significant extent (<10%) by peritoneal

TABLE 21-5  Dosing Adjustment of Cephalosporins in Patients with Renal Insufficiency


USUAL ADULT DOSING REGIMEN WITH
DOSING DOSING REGIMEN WITH RENAL INSUFFICIENCY DIALYSIS
CEPHALOSPORIN REGIMEN GFR <10 mL/min GFR 10-50 mL/min GFR 50-90 mL/min Hemodialysis CAPD
First Generation
Cefazolin 1 g q8h 0.5-1 g q24h 0.5-1 g q12h NC 0.5-1 g after 0.5 g q12h
Second Generation
Cefuroxime 1.5 g q8h 0.75g q24h 0.75 g q8-12h NC 0.75 g after 0.75 q24h
Cephamycins
Cefotetan 2 g q12h 1 g q24h 2 g q24h NC 1 g after 1 g q24h
Cefoxitin 2 g q6h 1 q q12h 2 g q12h 2 g q8h 1 g after 1 g q12h
Third Generation
Cefotaxime 2 g q8h 2 g q24h 2g q12h NC 1 g after 1 g q24h
Ceftazidime 2 g q8h 0.5 g q24h 2g q24h 2 g q12h 1 g after 0.5 q24h
Ceftriaxone 1 g q24h NC NC NC None NC
Fourth Generation
Cefepime 2 g q12h 0.5-1 g q24h 1g q24h NC 1 g after 0.5-1 g q24h
Cefpirome 2 g q12h 0.5 g q12h 1g q12h NC 1 g after 0.5 g three times
weekly
MRSA-Active*
Ceftobiprole 0.5 g q8h 0.25 g q12h 0.5 g q12h NC 0.25 g after 0.25 g q12h
Ceftaroline 0.6 g q12h 0.2 g q12h 0.3-0.4 g q1h NC 0.2 g after 0.2 g q12h
Oral: First Generation
Cefadroxil 500 mg q12h 500 mg q24h 500 mg q24h NC 500 mg after 500 mg q24h
Cephalexin 500 mg q6h 250 mg q12h 500 mg q12h NC 500 mg after 500 mg q12h
Oral: Second Generation
Cefprozil 500 mg q12h 250 mg q24h 500 mg q24h NC 500 mg after 250 mg q24h
Cefuroxime (axetil) 500 mg q8h 500 mg q24h 500 mg q12h NC 500 mg after 500 mg q24h
Oral: Third Generation
Cefdinir 300 mg q12h 300 mg q24h NC NC 300 mg after 300 mg q24h
Cefditoren 400 mg q12h 200 mg q24h 200 mg q12h NC None 200 mg q24h
Cefixime 400 mg q24h 200 mg q24h 300 mg q24h NC 300 mg after 200 mg q24h
Cefpodoxime 200 mg q12h 200 mg q24h NC NC 200 mg after 200 mg q24h
Ceftibuten 400 mg q24h 100 mg q24h 200 mg q24h NC 300 mg after 100 mg q24h
*Studies not available for ceftaroline.
CAPD, continuous ambulatory peritoneal dialysis; GFR, glomerular filtration rate; MRSA, methicillin-resistant Staphylococcus aureus; NC, no change.
287
dialysis, and additional dosing is not recommended after a peritoneal Use of the penicillin skin test to predict cephalosporin reactions is
dialysis session. Continuous venous hemofiltration (CVH) is fre- unreliable. In a study of almost 100 individuals who were given a
quently used in critically ill patients. Compounds eliminated by the cephalosporin and had a history of reaction to the penicillin determi-
kidneys and by hemodialysis are also removed by CVH. Most often, nants used in skin testing, only 1 patient had a reaction.163 The practical

Chapter 21  Cephalosporins


the efficiency of drug removal is thought to be similar to a creatinine clinical decision to use a cephalosporin in a patient with a prior history
clearance of 10 to 30 mL/min, and appropriate dosing modification is of reaction to either a penicillin or another cephalosporin should be
recommended.153 guided by the severity of the prior reaction and the cephalosporin to
be used. In patients with prior nonsevere, non–IgE-mediated reactions
ADVERSE REACTIONS AND to other β-lactams, use of a cephalosporin with a different side chain
TOXICITIES than the prior agent is considered safe.159,162 Although the risk of a
The safety profile of the cephalosporins as a class is generally favorable. similar reaction is slightly increased, the reactions are rarely severe.
The incidence of specific adverse reactions for these compounds is However, in the setting of a prior severe IgE-mediated reaction with
relatively similar among drugs within the class, with few exceptions another β-lactam compound, use of a cephalosporin is discouraged.
(Table 21-6). As with other β-lactam drugs, hypersensitivity reactions Immunologically mediated reactions to cephalosporins may mani-
are the most common adverse effect associated with cephalosporin fest as hematologic or renal toxicities. Eosinophilia is the most com-
therapy.154,155 The frequency of hypersensitivity reactions to cephalo- monly reported laboratory abnormality.164 Cytopenias associated with
sporins is less than that for penicillins. Various cutaneous rashes, often cephalosporin use are rare.165 Cytotoxic reactions from immuno­
associated with eosinophilia and occasionally with fever, occur in 1% globulin antibodies can rarely result in neutropenia, thrombocytope-
to 7% of patients receiving these drugs.156 More severe hypersensitivity nia, or anemia. A high incidence of neutropenia was observed after
reactions, such as serum sickness, anaphylaxis, or angioedema, occur prolonged therapy with cefepime for osteomyelitis but resolved after
very infrequently. These immunoglobulin E (IgE)-mediated reactions the drug was stopped.165 The incidence of hemolytic anemia is highest
are estimated to occur in fewer than 1 in 100,000 patients.157 However, with ceftriaxone.166 Although Coombs laboratory tests are reported to
there have been reports of a strong association between serum sickness be positive in a significant percentage of patients receiving many of the
in children and use of cefaclor.158 cephalosporins, these patients most often do not have hemolytic
Cross-reactivity among drugs from the cephalosporin class and anemia167 because this is a false-positive reaction related to cross-
other β-lactams has been extensively investigated. The risk appears to reactivity that occurs in up to 3% of patients. A similar cytotoxic reac-
depend on the similarity of side chains on the molecule to those on tion can result in renal damage from interstitial nephritis168; the
penicillins or other cephalosporins.159 For example, in patients with frequency of this reaction appears to be less than with drugs from the
allergy to amoxicillin, cross-reactivity was observed in 38% of those penicillin class.
receiving cefadroxil, which has a similar side chain, and in none of Nonimmunologic hematologic and renal toxicities have been
those receiving cefamandole, which has a different side chain.160 Recent reported with a similarly low frequency. Bleeding abnormalities have
estimates suggest a cross-reaction frequency of 1% or less.161 Cross- been reported with increased frequency related to two mechanisms.
reactivity with second-, third- and fourth-generation drugs has been Impaired adenosine diphosphate–induced platelet aggregation has
very low.161,162 been reported with moxalactam169 but not with other cephalosporins.
Another coagulopathy is specifically associated with the MTT side
chain present on cefamandole, cefotetan, cefoperazone, and moxalac-
tam.170 The MTT side chain can dissociate from the parent cephalo-
sporin and act as a competitive inhibitor of the vitamin K–dependent
TABLE 21-6  Potential Adverse Effects of carboxylase responsible for converting clotting factors II, VII, IX, and
Cephalosporins X to active forms.171 In addition, other cephalosporin side chains may
EFFECT TYPE SPECIFIC EFFECT FREQUENCY (%) interact with warfarin and increase inhibition of vitamin K 2,3-epoxide
Hypersensitivity Rash 1-3 reductase, which converts vitamin K to its active form.172,173 These reac-
Urticaria <1 tions can lead to prolongation of the prothrombin time and clinically
Serum sickness <1 significant bleeding. Patients with poor nutritional status, advanced
Anaphylaxis 0.01
age, or recent surgery on the gastrointestinal tract are at increased risk
for clinically significant bleeding.174-176 In addition, patients with renal
Gastrointestinal Diarrhea 1-19
failure may be at increased risk for bleeding because of the accumula-
Nausea/vomiting 1-6
tion of the side chain.177 Vitamin K administration rapidly reverses the
Transient transaminase 1-7 abnormality in 24 to 36 hours. The MTT side chain can also produce
elevation
a disulfiram-like reaction with ethanol ingestion that may persist for
Biliary sludge 20-46*
several days after antibiotic administration.13,178 The disulfiram reac-
Hematologic Eosinophilia 1-10 tion manifests with flushing, tachycardia, headache, sweating, nausea,
Neutropenia <1 vomiting, hypotension, confusion, or blurred vision. The reaction is
Thrombocytopenia <1-3 caused by a block in alcohol metabolism at the acetaldehyde step,
Hypoprothrombinemia <1 which results in the accumulation of acetaldehyde and subsequent
Impaired platelet <1 symptoms.
aggregation A variety of adverse reactions within the gastrointestinal tract have
Hemolytic anemia <1 been reported with variable frequency. Diarrhea is the most commonly
Renal Interstitial nephritis <1 reported side effect, with rates ranging from 1% to 20%.156 Upper gas-
Central nervous system Seizures <1 trointestinal symptoms occur much less frequently. Mild and transient
Encephalopathy <1 hepatic toxicity has been reported with most compounds from the class
False-positive laboratory result Coombs positive 3 and manifests as twofold to fourfold elevations in transaminase levels
Glucosuria Rare
in up to 7% of patients.179 Obstructive biliary toxicity has also been
reported with ceftriaxone.180,181 The high biliary ceftriaxone concentra-
Serum creatinine Rare
tions cause crystallization of a ceftriaxone-calcium salt and the clinical
Other Drug fever Rare
syndrome of biliary pseudolithiasis. This biliary abnormality has been
Disulfiram-like reaction† Rare reported most often in children who were receiving high ceftriaxone
Superinfection Rare doses and in patients with preexisting biliary abnormalities.182 The
Phlebitis Rare syndrome is reversible and in most reports has cleared within 10 to 60
*Ceftriaxone. days after discontinuation of the drug. It was recommended that cef-

Cephalosporins with thiomethyl tetrazole ring side chain. triaxone and calcium-containing products not be mixed in vials or
288
infusion lines for therapy in neonates younger than 28 days because of creatinine were reported in patients receiving cefoxitin or cephalothin
the risk of precipitation in their plasma. This warning was later in laboratories using the Jaffe technique.192
retracted as other cephalosporins had a similar low precipitation Because the cephalosporins have broad-spectrum activity, superin-
risk.183 fection or overgrowth of Candida in the gastrointestinal and vaginal
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

Adverse reactions in the nervous system are uncommon and are tracts can occur.193,194 Similarly, overgrowth of Clostridium difficile in
similar in nature to those reported with other β-lactams.184 The main the gastrointestinal tract with diarrhea and, less commonly, colitis has
mechanism of neurotoxicity is inhibition of γ-aminobutyric acid A.185 been associated with increased use of broad-spectrum cephalospo-
Encephalopathy and seizures have been reported primarily in patients rins.195,196 However, the incidence of these superinfections is similar to
with renal insufficiency who were receiving high doses of these rates reported with drugs from some other antibiotic classes.
drugs.185-187 Decreased protein binding and inhibition of the choroid
plexus pump occur with uremia and may contribute to enhanced toxic- CLINICAL USES OF SPECIFIC DRUGS
ity in patients with renal impairment. Local phlebitis reactions related The usual dosing regimens for adults and children for the various
to intravenous administration of the parenteral compounds have been cephalosporins by generation are listed in Table 21-7. The daily doses
reported with variable frequency, ranging from 1% to 5%.188 On the recommended to treat serious infections are also listed.
other hand, pain associated with intramuscular administration is
common with all of the parenteral compounds.189 The local discomfort First-Generation Cephalosporins
can be reduced by the use of 1% lidocaine in diluent. With the excep- The first-generation cephalosporins available in the United States are
tion of once-daily intramuscular ceftriaxone and cefepime, most par- cefazolin, cephalexin, and cefadroxil. The first-generation cephalospo-
enteral cephalosporins are administered by the intravenous route. rins have been extensively used as alternatives to penicillin for staphy-
The cephalosporins have not been studied extensively in pregnancy. lococcal and nonenterococcal streptococcal infections. Most commonly,
All drugs within the cephalosporin group are placed in pregnancy class these include skin and soft tissue infections.
B.190 All of the compounds are secreted to a small degree into breast Cefazolin is not metabolized and is eliminated more by glomerular
milk, but as a class they are considered safe for use in this situation. filtration than by tubular excretion.149 Its moderate protein binding
In addition to the false-positive Coombs test, laboratory abnormali- slows the glomerular filtration of the drug, resulting in a half-life of 1.5
ties in urine glucose and serum creatinine have been reported with to 2 hours, which allows 8- and 12-hourly dosing. With co-administration
certain cephalosporins. False-positive results on glucosuria tests per- of probenecid, cefazolin has been effective in skin and soft tissue infec-
formed with the copper reduction technique (Clinitest) have been tions with once-daily dosing.197 Cefazolin is still recommended in
reported with cefaclor, cefadroxil, cefamandole, cefonicid, cefotaxime, penicillin-allergic patients for more serious staphylococcal infections,
cefoxitin, and ceftazidime.191 Similarly, false increases in serum such as endocarditis, even though the drug is more readily hydrolyzed

TABLE 21-7  Dosing Regimens for Cephalosporins in Adults and Children


ADULT CHILDREN
CEPHALOSPORIN Usual Dose Severe Disease Usual Dose
First Generation
Cefazolin 0.5-1 g q8-12h 2 g q6-8h 12.5-33 mg/kg q6-8h
Second Generation
Cefuroxime 0.75-1.5 g q8h 1.5 g q8h 12.5-60 mg/kg q6-8h
Cephamycins
Cefotetan 1-2 g q12h 2-3 g q12h Not recommended
Cefoxitin 1-2 g q6h 2 g q4-6h 20-25 mg/kg q4-6h
Third Generation
Cefotaxime 1 g q8-12h 2 g q4-8h 25-30 mg/kg q4-6h
Ceftazidime 1 g q8-12h 2 g q8h 30-50 mg/kg q8h
Ceftriaxone 1 g q24h 2 g q12-24h 50-100 mg/kg q24h
Fourth Generation
Cefepime 1 g q12h 2 g q8-12h 50 mg/kg q8h
Cefpirome 1 g q12h 2 g q12h Not recommended
MRSA-Active
Ceftaroline 0.6 g q12h 0.6 g q8h Not recommended
Ceftobiprole 0.5 g q12h 0.5 g q8h Not recommended
Oral: First Generation
Cephalexin 250-500 mg qid 1 g qid 6.25-25 mg/kg qid
Cefadroxil 500 mg bid 1 g bid 15 mg/kg bid
Oral: Second Generation
Cefprozil 250-500 mg bid 500 mg bid 7.5-15 mg/kg bid
Cefuroxime (axetil) 250-500 mg bid 500 mg bid 10-15 mg/kg bid
Oral: Third Generation
Cefdinir 300 mg bid or 300 mg bid or 7 mg/kg bid or
600 mg qd 600 mg qd 14 mg/kg qd
Cefditoren 200-400 mg bid 400 mg bid Not recommended
Cefixime 200 mg bid or 400 mg bid 4 mg/kg bid or
400 mg qd 8 mg/kg qd
Cefpodoxime 200-400 mg bid 400 mg bid 5 mg/kg bid
Ceftibuten 400 mg qd 400 mg qd 9 mg/kg qd
MRSA, methicillin-resistant Staphylococcus aureus.
289
by staphylococcal β-lactamase than other first-generation cephalospo- cefprozil are the best agents for S. pneumoniae and are effective against
rins.198,199 Cefazolin is recommended as the prophylactic antibiotic of penicillin-susceptible isolates and most penicillin-intermediate strains.
choice for foreign-body implantation and for many clean and clean None of the oral second-generation cephalosporins provides optimal
contaminated surgical procedures in which there is a high risk of infec- therapy for penicillin-resistant pneumococci. For H. influenzae and M.

Chapter 21  Cephalosporins


tion.200 These include cardiac and vascular surgery, insertion of ortho- catarrhalis, cefuroxime axetil would be a better choice than cefprozil.
pedic devices, head and neck surgery that crosses the oropharyngeal Clinical and bacteriologic outcomes have demonstrated that 5- to
mucosal barrier, vaginal and abdominal hysterectomy, high-risk cesar- 10-day courses of therapy with the second-generation cephalosporins
ean section, and high-risk gastroduodenal and biliary tract procedures. are equivalent to or more effective than 10 days of therapy with penicil-
Because of its poor activity against Bacteroides spp., cefazolin alone is lin V for the treatment of group A β-hemolytic streptococcal pharyn-
not recommended for intra-abdominal procedures that involve the gitis.215,216 Cefuroxime axetil is also a recommended alternative to
intestine. doxycycline and penicillin for treatment of early Lyme disease.217
The oral first-generation cephalosporins, cephalexin and cefadroxil, The cephamycins, cefoxitin and cefotetan, have provided effective
have very high oral bioavailability. Cefadroxil has a slightly longer half- therapy for a variety of infections involving aerobic gram-negative
life than cephalexin, which allows twice-daily dosing instead of the and anaerobic organisms, especially B. fragilis. These include intra-
usual four times a day.107 These drugs provide appropriate outpatient abdominal, pelvic, and gynecologic infections; infected decubitus
therapy for many skin and soft tissue infections. However, they are not ulcers; diabetic foot infections; and mixed aerobic-anaerobic soft tissue
effective for animal bites and scratches involving Pasteurella multo- infections.218-220 Cefotetan has a half-life of 3 to 4 hours because of its
cida.201 The drugs are quite active against Streptococcus pyogenes and high protein binding and can be administered twice daily, compared
provide effective therapy in streptococcal pharyngitis.202,203 They have with every 6 hours for cefoxitin. Cefotetan contains the MTT side
poor activity against penicillin-resistant pneumococci, H. influenzae, chain, which increases its potential for hypoprothrombinemia and
and M. catarrhalis and are not recommended for sinusitis, otitis media, disulfiram reactions. In terms of antimicrobial activity, cefotetan is
or lower respiratory tract infections. The drugs are effective in uncom- more active than cefoxitin against gram-negative bacilli but is less
plicated urinary tract infections; however, they are less effective than active against non–B. fragilis members of the B. fragilis group.221,222 Of
trimethoprim-sulfamethoxazole or fluoroquinolones.204 more importance, as many as 15% to 20% of B. fragilis strains are
resistant to the various cephamycins, and drugs with better anaerobic
Second-Generation Cephalosporins activity should be used for empirical therapy of serious Bacteroides
The second-generation cephalosporins currently available in the infections.223 All of the cephamycins are active against N. gonorrhoeae,
United States include two true cephalosporins, cefprozil and cefurox- including penicillin-resistant strains. However, recommended therapy
ime, and two cephamycins, cefoxitin and cefotetan. The two groups of for infections with this organism is with ceftriaxone, which is effective
drugs have different spectra of antimicrobial activity and different as a single dose.224 However, resistance of N. gonorrhoeae to ceftriaxone
clinical uses. The true cephalosporins have increased activity against is starting to increase worldwide.225 Cefoxitin and cefotetan, in combi-
H. influenzae, M. catarrhalis, and Neisseria spp. and comparable activ- nation with doxycycline, provide effective therapy for pelvic inflamma-
ity against staphylococci and nonenterococcal streptococci. The cepha- tory disease.226 For antimicrobial prophylaxis during surgery, cefoxitin
mycins have inferior activity against staphylococci but enhanced and cefotetan are recommended over cefazolin only for colorectal pro-
antibacterial activity against certain Enterobacteriaceae. The cephamy- cedures and appendectomies.200 For elective colorectal surgery, cefoxi-
cins are especially noted for their activity against anaerobic bacteria, tin or cefotetan is still commonly administered even when an oral
especially B. fragilis. They also demonstrate good in vitro activity bowel preparation with erythromycin and neomycin is used.
against ESBL-producing strains of E. coli and K. pneumoniae. However,
their reliability in treating infections caused by ESBL-producing strains Third-Generation Cephalosporins
has not been proven. Third-generation cephalosporins available in the United States are
Because of their activity against S. pneumoniae, H. influenzae, and cefdinir, cefditoren, cefixime, cefotaxime, cefpodoxime, ceftazidime,
M. catarrhalis, the true second-generation cephalosporins, such as ceftibuten, and ceftriaxone. The third-generation cephalosporins are
cefuroxime, have been used extensively for treatment of various respi- major drugs for the treatment of many important infections because
ratory tract infections in hospitalized patients.205 Cefuroxime can also of their high antibacterial potency, wide spectrum of activity, low
be used to treat meningitis caused by penicillin-susceptible pneumo- potential for toxicity, and favorable pharmacokinetics (e.g., enhanced
cocci, H. influenzae, or Neisseria meningitidis.206 However, its use has drug concentrations in the CSF). They have been especially useful in
been largely replaced by third-generation cephalosporins, which result infections resulting from gram-negative bacilli that are resistant to
in faster eradication of bacteria from the CSF.207 Cefuroxime was in other β-lactam antibiotics. However, their superior activity against the
the past one of the recommended agents for empirical therapy of Enterobacteriaceae is being challenged by the increasing frequency
community-acquired pneumonia (CAP) in hospitalized patients.208 of organisms with β-lactamase–mediated resistance. New AmpC
Although cefuroxime has good activity against penicillin-susceptible β-lactamases, ESBLs, and carbapenemases, which inactivate third-
and penicillin-intermediate strains of S. pneumoniae, its activity against generation cephalosporins, present a distinct threat to the continued
most penicillin-resistant strains is suboptimal. In an observational utility of these agents.
study of 844 patients with pneumococcal bacteremia, mostly resulting Cefotaxime, ceftriaxone, and ceftazidime are the major parenteral
from pneumonia, resistance to cefuroxime was associated with signifi- third-generation cephalosporins in clinical use for the treatment of
cantly greater mortality.209 The latest recommendations on CAP list nosocomial infections caused by susceptible gram-negative bacilli.
ceftriaxone or cefotaxime for initial empirical therapy.210 Cefuroxime Cefotaxime and ceftriaxone are also two of the most potent cephalo-
provides effective therapy for other serious infections caused by sus- sporins against penicillin-resistant pneumococci. Because of its high
ceptible pathogens, including skin and soft tissue infections, epiglot- protein binding, ceftriaxone has the longest half-life and is usually
titis, complicated sinusitis, and gynecologic infections.211,212 Cefuroxime administered once daily. Ceftazidime is dosed two or three times daily,
should not be considered for empirical therapy of nosocomial pneu- and effective dosing of cefotaxime, which has the shortest half-life, has
monia or other nosocomial infections because of its poor activity varied from every 4 hours to twice daily. Ceftazidime is usually reserved
against most strains of Enterobacter, Citrobacter, Serratia, Morganella, for infections that are likely to involve P. aeruginosa.
and P. aeruginosa. Monotherapy with cefotaxime or ceftriaxone has provided effec-
The oral second-generation cephalosporins, including cefuroxime tive treatment for a variety of nosocomial infections caused by sus­
axetil and cefprozil, are effective for treatment of a variety of mild-to- ceptible gram-negative bacilli, including complicated skin and soft
moderate community-acquired infections. Double-tap studies in acute tissue infections, prosthetic joint infections, pneumonia, complicated
otitis media and acute maxillary sinusitis demonstrated that bacterial urinary tract infections, and intra-abdominal infections such as
eradication was related to the drug’s ability to produce serum concen- peritonitis.227-231 However, cephalosporin monotherapy for infections
trations exceeding the MIC of the infecting pathogen for 40% to 50% caused primarily by Enterobacter, Citrobacter, and Serratia spp. can
of the dosing interval.213,214 In this regard, cefuroxime axetil and be complicated by the emergence of stably derepressed resistant
290
mutants.84,232 Combination antimicrobial therapy may be beneficial in intramuscularly is highly active against N. gonorrhoeae, including
reducing this emergence of resistance, which results from increased penicillin- and quinolone-resistant strains. It is the drug of choice for
chromosomal β-lactamase production. Organisms containing ESBLs all forms of gonococcal infection and is used in combination with a
have also been observed to result in failed therapy with cephalospo- single oral dose of azithromycin or 7 days of oral doxycycline.224 A
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

rins, even when the organism was judged susceptible in laboratory single oral dose of cefixime is also highly effective for uncomplicated
tests.91 The carbapenems are the recommended drugs for these ESBL- gonococcal infections of the cervix, urethra, and rectum.254,255 However,
producing strains. because of increasing resistance, it is no longer recommended as first-
Cefotaxime and ceftriaxone have provided effective therapy for line therapy.256 Single-dose intramuscular ceftriaxone is recommended
meningitis caused by a variety of different bacteria.233-236 They are the therapy for chancroid.224 Ceftriaxone and cefotaxime are recom-
drugs of choice for meningitis caused by H. influenzae and various mended therapy for treatment of early Lyme disease in patients with
Enterobacteriaceae.237 Cefotaxime and ceftriaxone also provide effec- neurologic involvement or third-degree atrioventricular heart block.257
tive therapy for meningitis caused by N. meningitidis and against pneu- These drugs are also recommended in Lyme disease patients with both
mococci that have MICs of 1.0 µg/mL or less. Organisms with higher arthritis and objective evidence of neurologic disease and for those
MICs have resulted in failed monotherapy with these cephalosporins. with late neurologic disease affecting the central or peripheral nervous
Therefore, empirical therapy with cefotaxime or ceftriaxone is com- system. Ceftriaxone is considered as alternative therapy in penicillin-
bined with vancomycin (with or without rifampin) until the laboratory allergic patients with syphilis.224,258 Ceftriaxone is a recommended
determines the susceptibility of the pneumococcal isolate.237 If the alternative therapy for typhoid fever and for severe infections caused
organism is susceptible to cefotaxime or ceftriaxone, the vancomycin by Shigella spp. or by non-typhi spp. of Salmonella.259 Third-generation
(and rifampin) can be discontinued. Treatment of meningitis requires cephalosporins have provided effective therapy for focal Salmonella
maximal doses of these cephalosporins, such as 2 g every 12 hours in infections, brain abscess caused by gram-negative bacilli, and endocar-
adults and 50 mg/kg twice daily or 100 mg/kg once daily in children ditis caused by fastidious gram-negative coccobacilli.260,261 Single doses
for ceftriaxone, and 2 g every 4 to 6 hours in adults or 100 to 150 mg/ of ceftriaxone are highly effective in eradicating nasopharyngeal car-
kg every 6 to 8 hours in children for cefotaxime. riage of N. meningitidis.262,263 The long half-life of ceftriaxone, which
Cefotaxime and ceftriaxone continue to be active against most bac- allows for once-daily dosing, has enhanced its use in the outpatient
teria producing CAP. In a large observational study of pneumococcal setting for both streptococcal and staphylococcal infections. Ceftriax-
bacteremia, resistance to cefotaxime and ceftriaxone was not associ- one has been effective for the outpatient treatment of staphylococcal
ated with higher mortality.209 Cefotaxime and ceftriaxone were also and streptococcal skin and soft tissue infections, including osteomyeli-
effective in treating patients with nonmeningeal pneumococcal infec- tis and prosthetic joint infection.231,264,265 The drug is also effective as
tions, mostly pneumonia, caused by strains with MICs as high as 2 µg/ monotherapy for the outpatient treatment of nonenterococcal strepto-
mL.238 The CLSI has created higher susceptibility and resistance break- coccal endocarditis.266
points for pneumococci causing nonmeningeal infections than for S.
pneumoniae causing meningitis.239 As a result of these changes, cefo- Fourth-Generation Cephalosporins
taxime and ceftriaxone are considered active against most penicillin- Cefepime is the only fourth-generation cephalosporin available in the
resistant pneumococci and are recommended, in combination with a United States. The fourth-generation cephalosporins have the widest
macrolide, for empirical therapy for CAP requiring hospitalization.210 spectrum of all the cephalosporins. They have enhanced activity
Single intramuscular doses of ceftriaxone are also highly effective in against certain gram-negative bacilli, such as Enterobacter, Citrobacter,
eradicating H. influenzae and penicillin-susceptible strains of S. pneu- and Serratia spp. These drugs are zwitterions, which cross the outer
moniae from middle ear fluid.240 However, three daily doses of ceftri- membrane of gram-negative bacilli more rapidly than other cephalo-
axone were required in one study to eradicate penicillin-resistant sporins. They are also less susceptible to inactivation by AmpC
pneumococci.241 β-lactamases. As a result, about 75% to 80% of the Enterobacteriaceae
The oral third-generation cephalosporins, which include cefdinir, resistant to ceftazidime are susceptible to the fourth-generation drugs.
cefditoren pivoxil, cefixime, cefpodoxime proxetil, and ceftibuten, are They are active against P. aeruginosa and, unlike ceftazidime, maintain
approved for oral therapy of mild-to-moderate respiratory infections, good potency against gram-positive cocci. Only two fourth-generation
such as otitis media, sinusitis, and acute exacerbations of chronic bron- agents have been developed so far. Cefepime has a slightly longer half-
chitis. These drugs have very potent activity against H. influenzae, but life than ceftazidime and is usually administered twice daily, although
their activity against pneumococci is more variable.242-244 Cefdinir, 8-hour dosing is recommended for documented P. aeruginosa infec-
cefditoren, and cefpodoxime have activity similar to that of cefuroxime tions. Cefpirome, which is not available in the United States, has a
or cefprozil and are active against penicillin-susceptible and most pharmacokinetic profile very similar to that of cefepime. Both of these
penicillin-intermediate strains of S. pneumoniae. Cefixime is active agents maintain excellent activity against methicillin-susceptible S.
only against penicillin-susceptible strains, and ceftibuten is marginal aureus, S. pneumoniae (including most penicillin-resistant strains),
even for penicillin-susceptible pneumococci. Short courses of most of and other streptococci.267,268
these drugs have also provided equivalent rates of eradication in group At some hospitals, the fourth-generation agents have largely
A β-hemolytic streptococcal pharyngitis.245-247 Their increased potency replaced third-generation cephalosporins for treatment of serious
over other oral cephalosporins for E. coli, K. pneumoniae, and Entero- gram-negative bacillary infections. They have proved to be effective in
bacter, Citrobacter, and Serratia spp. enhances their utility for treat- a variety of serious gram-negative infections, such as bacteremia, pneu-
ment of complicated urinary tract infections.248 monia, skin and soft tissue infections, prosthetic joint infections, and
Ceftazidime is the third-generation cephalosporin used for serious complicated urinary tract infections.231,269-271 With continuous infusion
infections in which P. aeruginosa is documented or highly likely. It is of cefepime, optimal efficacy was observed when serum concentrations
one of the recommended drugs, either alone or in combination with were at least fourfold greater than the MIC.272 Comparative trials with
an aminoglycoside, for initial empirical management of febrile neutro- ceftazidime or other agents have generally demonstrated equivalent
penia.249 However, ESBLs and AmpC β-lactamases have reduced the efficacy.273-276 Still, cefepime has demonstrated efficacy against organ-
utility of ceftazidime for monotherapy. Continuous infusion of ceftazi- isms with reduced susceptibility or resistance to ceftazidime.277
dime has been used to increase trough concentrations, but trials of Cefepime is recommended as monotherapy or in combination with an
intermittent versus continuous administration have only rarely dem- aminoglycoside for initial empirical therapy in febrile neutropenic
onstrated any significant difference in efficacy.250-252 Ceftazidime has patients. The drug is one of the recommended agents for the empirical
been effective for the treatment of acute exacerbations of chronic pul- treatment of severe CAP when P. aeruginosa is suspected. The drug’s
monary infections in patients with cystic fibrosis. The drug penetrates activity against pneumococci is similar to that of ceftriaxone in patients
into CSF and is one of two cephalosporins as treatment of choice for with CAP requiring hospitalization.278,279 The drug penetrates well into
meningitis caused by P. aeruginosa.253 the CSF and produces outcomes similar to those of cefotaxime in acute
Third-generation cephalosporins have also become established bacterial meningitis.280,281 However, it is not approved for treatment of
therapy for a variety of specific infections. Ceftriaxone 250 to 500 mg meningitis in the United States. Although emergence of resistance
291
during therapy may be less of a problem with the fourth-generation daptomycin-nonsusceptible MRSA, resulting in enhanced killing and
cephalosporins, it does occur. Many of the new ESBLs and carbapen- improved clinical outcome.295,296 Little is known about the CSF penetra-
emases can inactivate these fourth-generation drugs. tion of ceftobiprole and ceftaroline in humans or whether the enhanced
A recent meta-analysis of cefepime versus other β-lactam drugs in activity of these drugs against pneumococci will be useful in treating

Chapter 21  Cephalosporins


the treatment of febrile neutropenia and other serious infections meningitis. These drugs may also become useful agents for treating
reported that all-cause mortality with cefepime was higher than with severe enterococcal infections in penicillin-allergic patients.297
other cephalosporins or with a β-lactam/β-lactamase inhibitor combi-
nation.282,283 An additional report suggested that the difference in mor- Cephalosporins Plus β-Lactamase
tality in patients with febrile neutropenia was caused by greater Inhibitors
progression of underlying disease in the cefepime arm.284 A meta- Because of increasing resistance to cephalosporins, more recent efforts
analysis by the Center for Drug Evaluation of the U.S. Food and Drug have been focused on combining some cephalosporins with β-lactamase
Administration using a larger database of both published and nonpub- inhibitors. The current preparation being studied combines ceftazi-
lished comparative studies did not find a significant increase in mortal- dime and avibactam at a 4 : 1 ratio, ceftaroline and avibactam at a 1 : 1
ity in cefepime-treated patients, compared with those treated with ratio, and ceftolozane and tazobactam at a 2 : 1 ratio.18 The addition of
other antibacterials.285 avibactam to ceftazidime completely restores full susceptibility (MIC
≤ 1 mg/L) for a collection of almost 300 ceftazidime-resistant strains
MRSA-Active Cephalosporins producing ESBLs, hyperproducing AmpCs, or both.298 For P. aerugi-
The MRSA-active cephalosporins have unique activity compared with nosa, avibactam reduces the MIC90 fourfold and produces MICs less
other cephalosporins. They have excellent activity again MRSA and than or equal to 8 mg/L for 60% of multidrug-resistant strains.299 Avi-
against ampicillin-susceptible strains of E. faecalis.59,62 They also exhibit bactam has minimal effect on ceftazidime MICs for Acinetobacter bau-
potent activity against pneumococci with multiple mutations in genes manii and most anaerobic bacteria.300,301 At concentrations less than or
encoding PBPs 1A, 2B, and 2X. MIC90 values that were as high as 4 equal to 4 mg/L, avibactam also protects ceftaroline against Enterobac-
and 8 µg/mL to the most active third-generation cephalosporins were teriaceae with ESBLs, AmpCs, and most KPC carbapenemases.302,303
fourfold to eightfold lower with the MRSA-active cephalosporins.286,287 However, strains with metallo-β-lactamases remain resistant. Avibac-
On the other hand, the activity of these drugs against gram-negative tam is eliminated primarily in the urine, with a half-life of approxi-
bacilli is similar to that of many of the third-generation cephalospo- mately 2 hours, which is similar to the half-lives of ceftazidime and
rins. The first approved drug in this group (in a few countries, but not ceftaroline.301 So far, the combination of avibactam and ceftazidime has
the United States) was ceftobiprole medocaril, the prodrug for ceftobi- resulted in comparable efficacy to carbapenems in complicated urinary
prole. The U.S.-approved MRSA-active cephalosporin is ceftaroline tract infections and in complicated intra-abdominal infections.304,305
fosamil, the prodrug for ceftaroline. Ceftobiprole is more resistant than Tazobactam is the β-lactamase inhibitor that is being combined
ceftaroline to inactivation by AmpC β-lactamase.81 Ceftobiprole also with ceftolozane, a new cephalosporin with enhanced activity against
has activity against strains of P. aeruginosa that is similar to that of strains of P. aeruginosa. The MICs of ceftolozane against P. aeruginosa
ceftazidime. However, the maximal doses of ceftobiprole studied so far are 4- to 16-fold lower than ceftazidime.306,307 The drug also appears to
are fourfold lower than those of ceftazidime. have earlier in vitro killing and more rapid in vivo killing than ceftazi-
The MRSA-active cephalosporins have exhibited comparable effi- dime.308,309 Although tazobactam has a minimal effect on ceftolozane’s
cacy to vancomycin plus a broad-spectrum β-lactam in patients with activity against P. aeruginosa, it does enhance its activity against other
complicated skin and skin-structure infections.288-290 The results with gram-negative bacilli producing various β-lactamases. At a concentra-
ceftobiprole and ceftaroline were similar to those obtained with van- tion of ceftolozane and tazobactam of 8 and 4 mg/L, respectively, 70%
comycin in the large number of patients with MRSA infections. to 76% of Enterobacter strains that are AmpC hyperproducers or pro-
Studies with ceftaroline and ceftobiprole have shown efficacy in ducing ESBLs are susceptible to the drug combination.310 Isolates with
hospitalized patients with CAP similar to that of comparators.291,292 KPC or other carbapenemases remain resistant. The half-life of tazo-
Ceftaroline has been very effective in small groups of patients with bactam is about 1 hour, which is 2.5 times shorter than the half-life of
complicated MRSA bacteremia with or without endocarditis.293,294 Cef- ceftolozane.128 This combination is currently being evaluated in com-
taroline also increases the membrane binding of daptomycin against parative clinical trials.

Key References 53. Pfaller MA, Sader HS, Fritsche TR, et al. Antimicrobial
activity of cefepime tested against ceftazidime-resistant
112. Barriere SL, Flaherty JR. Third generation cephalosporins:
a critical evaluation. Clin Pharm. 1984;3:351-373.
gram-negative clinical strains from North American Hos- 133. Cherubin CE, Eng RHK, Norrby R, et al. Penetration of
The complete reference list is available online at Expert Consult.
pitals: report from the SENTRY Antimicrobial Surveillance newer cephalosporins into cerebrospinal fluid. Rev Infect
Program (1998-2004). Diagn Microbiol Infect Dis. 2006;56: Dis. 1989;11:526-548.
1. Bo G. Giuseppe Brotzu and the discovery of cephalospo-
63-68. 145. Andes D, Craig WA. Pharmacokinetics and pharmacody-
rins. Clin Microbiol Infect. 2000;6(suppl 3):6-9.
63. Ge Y, Biek D, Talbot GH, et al. In vitro profiling of ceftaro- namics of antibiotics in meningitis. Infect Dis Clin North
5. Neu HC. β-Lactam antibiotics: structural relationships
line against a collection of recent bacterial clinical isolates Am. 1999;13:595-617.
affecting in vitro activity and pharmacologic properties.
from across the United States. Antimicrob Agents Che- 159. Pichichero ME. A review of evidence supporting the
Rev Infect Dis. 1986;8(suppl 3):S237-S259.
mother. 2008;52:3398-3407. American Academy of Pediatrics recommendation for pre-
14. Hanaki H, Akagi H, Nomura S, et al. Structure-activity
74. Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev. scribing cephalosporin antibiotics for penicillin-allergic
relationships of cephalosporin derivatives against
2009;22:161-182. patients. Pediatrics. 2005;115:1048-1057.
methicillin-resistant Staphylococcus aureus and Enterococ-
78. Bush K. Proliferation and significance of clinically relevant 162. Pichichero ME, Casey JR. Safe use of selected cephalospo-
cus faecalis. J Antibiot. 1996;49:402-404.
β-lactamases. Ann N Y Acad Sci. 2013;1277:84-90. rins in penicillin-allergic patients: a meta-analysis. Otolar-
18. Shlaes DM. New β-lactam-β-lactamase inhibitor combina-
92. Kahlmeter G. Breakpoints for intravenously used cephalo- yngol Head Neck Surg. 2007;136:340-347.
tions in clinical development. Ann N Y Acad Sci. 2013;
sporins in Enterobacteriaceae: EUCAST and CLSI break- 173. Baillargeon J, Holmes HM, Lin YL, et al. Concurrent use
1277:105-114.
points. Clin Microbiol Infect. 2008;14(suppl 1):169-174. of warfarin and antibiotics and the risk of bleeding in older
22. Spratt BG. Distinct penicillin-binding protein involved in
93. Dudley MN, Ambrose PG, Bhavnani SM, et al. Background adults. Amer J Med. 2012;125:183-189.
the division, elongation and shape of Escherichia coli K12.
and rationale for revised Clinical and Laboratory Standards 183. Steadman E, Raisch DW, Bennett CL, et al. Evaluation
Proc Natl Acad Sci U S A. 1975;72:2999-3003.
Institute interpretive criteria (breakpoints) for Enterobac- of a potential clinical interaction between ceftriaxone
26. Craig WA. Interrelationship between pharmacokinetics
teriaceae and Pseudomonas aeruginosa: 1. Cephalosporins and calcium. Antimicrob Agents Chemother. 2010;54:
and pharmacodynamics in determining dosage regimens
and aztreonam. Clin Infect Dis. 2013;56:1301-1309. 1534-1540.
for broad-spectrum cephalosporins. Diagn Microbiol Infect
95. Lee NY, Lee CC, Huang WH, et al. Cefepime therapy 185. Grill MF, Maganti R. Cephalosporin-induced neurotoxic-
Dis. 1995;22:89-96.
for monomicrobial bacteremia caused by cefepime- ity: clinical manifestations, potential pathogenic mecha-
29. Bergeron MG, Bausch JL, Barza M, et al. Bactericidal activ-
susceptible extended-spectrum beta-lactamase-producing nisms, and the role of electroencephalographic monitoring.
ity and pharmacology of cefazolin. Antimicrob Agents Che-
Enterobacteriaceae: MIC matters. Clin Infect Dis. 2013;56: Ann Pharmacol. 2008;42:1843-1850.
mother. 1973;4:396-401.
488-495. 200. Bratzler DW, Dellinger EP, Olsen KM, et al. Clinical prac-
38. Cleeland R, Squires E. Antimicrobial activity of ceftriax-
99. Masuda N, Sakagawa E, Ohya S, et al. Substrate specificities tice guideline for antimicrobial prophylaxis in surgery. Am
one, a review. Am J Med. 1984;77:3-11.
of MexAB-OprM, MexCD-OprJ, and MexXY-OprM efflux J Health-Syst Pharm. 2013;70:195-283.
52. Karlowsky JA, Jones MY, Hayfield DC, et al. Ceftriaxone
pumps in Pseudomonas aeruginosa. Antimicrob Agents 207. Schaad UB, Suter S, Gianella-Borradori A, et al. A com-
activity against gram-positive and gram-negative patho-
Chemother. 2000;44:3322-3327. parison of ceftriaxone and cefuroxime for the treatment of
gens isolated in US clinical microbiology laboratories from
110. Brogard JM, Conte E. Pharmacokinetics of the new cepha- bacterial meningitis in children. N Engl J Med. 1990;322:
1996 to 2000: results from The Surveillance Network (TSN)
losporins. Antibiot Chemother. 1982;31:145-210. 141-147.
Database-USA. Int J Antimicrob Agents. 2002;19:413-426.
292
210. Mandell LA, Wunderink RG, Anzueto A, et al. Infectious 253. Fong IW, Tompkins KB. Review of Pseudomonas aerugi- with ceftaroline salvage therapy. J Antimicrob Chemother.
Diseases Society of America/American Thoracic Society nosa meningitis with special emphasis on treatment with 2012;67:1267-1270.
consensus guidelines on the management of community- ceftazidime. Rev Infect Dis. 1985;7:604-612. 295. Werth BJ, Sakoulas G, Rose WE, et al. Ceftaroline increases
acquired pneumonia in adults. Clin Infect Dis. 2007;44 273. Hoepelman AI, Kieft H, Aoun M, et al. International com- membrane binding and enhances the activity of daptomy-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

(suppl 2):S27-S72. parative study of cefepime and ceftazidime in the treatment cin against daptomycin-nonsusceptible vancomycin-
224. Centers for Disease Control and Prevention. Sexually of serious bacterial infections. J Antimicrob Chemother. intermediate Staphylococcus aureus in a pharmacokinetic/
transmitted diseases treatment guidelines, 2010. MMWR 1993;32(suppl B):175-186. pharmacodynamic model. Antimicrob Agents Chemother.
Morb Mortal Wkly Rep. 2010;51(RR-12):1-110. 275. Cordonnier C, Herbrecht R, Pico JL, et al. Cefepime/ 2013;57:66-73.
231. Osmon DR, Berbari EF, Berendt AR, et al. Diagnosis and amikacin versus ceftazidime/amikacin as empirical therapy 298. Lagace-Wiens PR, Tailor F, Simner P, et al. Activity of
management of prosthetic joint infection: clinical practice for febrile episodes in neutropenic patients: a comparative NXL104 in combination with beta-lactams against geneti-
guidelines by the Infectious Diseases Society of America. study. Clin Infect Dis. 1997;24:41-51. cally characterized Escherichia coli and Klebsiella pneu-
Clin Infect Dis. 2013;56:e1-e25. 283. Yahav D, Paul M, Fraser A, et al. Efficacy and safety of moniae isolates producing class A extended-spectrum
233. Congeni BL, Bradley J, Hammerschlag MR. Safety and effi- cefepime: a systematic review and meta-analysis. Lancet beta-lactamases and class C beta-lactamases. Antimicrob
cacy of once daily ceftriaxone for the treatment of bacterial Infect Dis. 2007;7:338-348. Agents Chemother. 2011;55:2434-2437.
meningitis. Pediatr Infect Dis. 1986;5:293-297. 285. Kim PW, Wu YT, Cooper B, et al. Meta-analysis of a pos- 301. Zhanel GG, Lawson CD, Adam H, et al. Ceftazidime-
237. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guide- sible signal of increased mortality associated with cefepime avibactam: a novel cephalosporin/β-lactamase inhibitor
lines for management of bacterial meningitis. Clin Infect use. Clin Infect Dis. 2010;51:381-389. combination. Drugs. 2013;73159-73177.
Dis. 2004;39:1267-1284. 290. Corey GR, Wilcox M, Talbot GH, et al. Integrated analysis 302. Mushtaq S, Warner M, Williams G, et al. Activity of che-
238. Pallares R, Capdevila Q, Linares J, et al. The effect of cepha- of CANVAS 1 and 2: phase 3, multicenter, randomized, querboard combinations of ceftaroline and NXL104 versus
losporin resistance on mortality in adult patients with double-blind studies to evaluate the safety and efficacy of beta-lactamase-producing Enterobacteriaceae. J Antimi-
nonmeningeal systemic pneumococcal infections. Am J ceftaroline versus vancomycin plus aztreonam in compli- crob Chemother. 2010;65:1428-1432.
Med. 2002;113:120-126. cated skin and skin-structure infections. Clin Infect Dis. 307. Sader HS, Rhomberg PR, Farrell DJ, Jones RN. Antimicro-
241. Leibovitz E, Piglansky L, Raiz S, et al. Bacteriologic and 2010;51:641-650. bial activity of CXA-101, a novel cephalosporin testing
clinical efficacy of one day vs. three day intramuscular cef- 292. File TM Jr, Low DE, Eckburg PB, et al. Integrated analysis combination with tazobactam against Enterobacteriaceae,
triaxone for treatment of nonresponsive acute otitis media of FOCUS 1 and FOCUS 2: randomized, double-blinded, Pseudomonas aeruginosa, and Bacteroides fragilis strains
in children. Pediatr Infect Dis J. 2000;19:1040-1050. multicenter phase 3 trials of the efficacy and safety of having various resistance phenotypes. Antimicrob Agents
249. Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical practice ceftaroline fosamil versus ceftriaxone in patients with Chemother. 2011;55:2390-2394.
guideline for the use of antimicrobial agents in neutrope- community-acquired pneumonia. Clin Infect Dis. 2010; 310. Livermore DM, Mushtaq S, Ge Y. Chequerboard titration
nic patients with cancer: 2010 update by the Infectious 51:1395-1405. of cephalosporin CXA-101 (FR264205) and tazobactam
Diseases Society of America. Clin Infect Dis. 2011;52: 293. Ho TT, Cadena J, Childs LM, et al. Methicillin-resistant versus beta-lactamase-producing Enterobacteriaceae.
e56-e93. Staphylococcus aureus bacteremia and endocarditis treated J Antimicrob Chemother. 2010;65:1972-1974.
292.e1
References glycopeptides, and linezolid. Infect Dis Clin North Am.
2003;17:479-501.
Program (1998-2004). Diagn Microbiol Infect Dis. 2006;56:
63-68.
1. Bo G. Giuseppe Brotzu and the discovery of cephalospo-
29. Bergeron MG, Bausch JL, Barza M, et al. Bactericidal activ- 54. Spangler SK, Jacobs MR, Pankuch GA, et al. Susceptibility
rins. Clin Microbiol Infect. 2000;6(suppl 3):6-9.
ity and pharmacology of cefazolin. Antimicrob Agents Che- of 170 penicillin-susceptible and penicillin-resistant pneu-
2. Sykes RB. From moulds to drugs. Clin Microbiol Infect.

Chapter 21  Cephalosporins


mother. 1973;4:396-401. mococci to six oral cephalosporins, four quinolones,
2000;6(suppl 3):10-12.
30. Sabath LD, Wilcox C, Garner C, et al. In vitro activity of desacetyl cefotaxime, Ro 23-9424 and RP 67829. J Antimi-
3. Rolinson GN. The influence of 6-aminoenicillanic acid on
cefazolin against recent clinical bacterial isolates. J Infect crob Chemother. 1993;31:273-280.
antibiotic development. J Antimicrob Chemother. 1988;22:
Dis. 1973;128(suppl):S320-S326. 55. Thornsberry C, Ogilvie P, Kahn J, et al. Surveillance of
5-14.
31. Bill NJ, Washington JA. Comparison of in vitro activity of antimicrobial resistance in Streptococcus pneumoniae, Hae-
4. Neu HC. Structure-activity relations of new β-lactam com-
cephalexin, cephradine and cefaclor. Antimicrob Agents mophilus influenzae, and Moraxella catarrhalis in the
pounds and in vitro activity against common bacteria. Rev
Chemother. 1977;11:470-474. United States in 1996-1997 respiratory season. Diagn
Infect Dis. 1983;5(suppl 2):S319-S336.
32. Sanders CC. In vitro studies with cefaclor, a new oral Microbial Infect Dis. 1997;29:249-257.
5. Neu HC. β-Lactam antibiotics: structural relationships
cephalosporin. Antimicrob Agents Chemother. 1977;12: 56. Doern GV, Jones RN, Pfaller MA, et al. Haemophilus influ-
affecting in vitro activity and pharmacologic properties.
490-497. enzae and Moraxella catarrhalis from patients with
Rev Infect Dis. 1986;8(suppl 3):S237-S259.
33. Hartstein Al, Patrick KE, Jones SR, et al. Comparison of community-acquired respiratory tract infections: anti­
6. Onishi HR, Daoust DR, Zimmerman SB, et al. Cefoxitin,
pharmacologic antimicrobial properties of cephadroxil and microbial susceptibility patterns from the SENTRY Anti-
a semisynthetic cephamycin antibiotic: resistance to
cephalexin. Antimicrob Agents Chemother. 1977;12:93-97. microbial Surveillance Program (United States and
β-lactamase inactivation. Antimicrob Agents Chemother.
34. Birnbaum J, Stapley EO, Miller AK, et al. Cefoxitin, a semi- Canada, 1997). Antimicrob Agents Chemother. 1999;43:
1974;5:38-48.
synthetic cephamycin: a microbiologic overview. J Antimi- 385-389.
7. Stapley EO, Birnbaum J. Chemistry and microbiological
crob Chemother. 1978;4:15-32. 57. Hoban D, Waites K, Felmingham D. Antimicrobial suscep-
properties of the cephamycins. In: Salton MRJ, Shockman
35. Eickhoff TC, Ehret JM. In vitro comparison of cefoxitin, tibility of community-acquired respiratory tract pathogens
GD, eds. Beta-Lactam Antibiotics. New York: Academic
cefamandole, cephalexin, and cephalothin. Antimicrob in North America in 1999-2000: findings of the PROTEKT
Press; 1981:327-351.
Agents Chemother. 1976;9:994-999. surveillance study. Diagn Microbiol Infect Dis. 2003;45:
8. Dunn GL. Ceftizoxime and other third generation cepha-
36. Greenwood D, Pearson N, Eley A, et al. Comparative 251-259.
losporins: structure activity relationships. J Antimicrob
in-vitro activities of cefotaxime and ceftizoxime (FK 749): 58. Karlowsky JA, Critchley IA, Blosser-Middleton RS, et al.
Chemother. 1982;10(suppl C):1-10.
new cephalosporins with exceptional potency. Antimicrob Antimicrobial surveillance of Haemophilus influenzae in
9. Naito T, Aburki S, Kamachi H, et al. Synthesis and struc-
Agents Chemother. 1980;17:397-401. the United States during 2000-2001 leads to detection of
ture activity relationship of a new series of cephalosporins,
37. Acuna G, Johnston J, Young LS, et al. In-vitro studies clonal dissemination of a beta-lactamase-negative and
BMY-28142 and related compounds. J Antibiot. 1986;39:
with ceftazidime against aerobic gram-negative bacilli and ampicillin-resistant strain. J Clin Microbiol. 2002;40:
1092-1107.
Bacteroides fragilis group. J Antimicrob Chemother. 1981; 1063-1066.
10. Toda A, Ohki H, Yamanaka T, et al. Synthesis and SAR of
8(suppl B):83-89. 59. Jones RN, Deshpande LM, Mutnick AH, et al. In vitro
novel parenteral anti-pseudomonal cephalosporins: dis-
38. Cleeland R, Squires E. Antimicrobial activity of ceftriax- evaluation of BAL9141, a novel parenteral cephalosporin
covery of FR264205. Bioorg Med Chem Lett. 2008;18:
one, a review. Am J Med. 1984;77:3-11. active against oxacillin-resistant staphylococci. J Antimi-
4849-4852.
39. Ayers LW, Jones RN, Barry AL, et al. Cefotetan, a new crob Chemother. 2002;50:915-932.
11. Turnidge JD, Craig WA. Beta-lactam pharmacology in liver
cephamycin: comparison of in vitro antimicrobial activity 60. Bogdanovich T, Clark C, Ednie L, et al. Activities of cefto-
disease. J Antimicrob Chemother. 1983;11:499-501.
with other cephems, beta-lactamase stability, and prelimi- biprole, a novel broad-spectrum cephalosporin, against
12. Bechtold H, Andrassy K, Jahnchen E, et al. Evidence for
nary recommendations for disk diffusion testing. Antimi- Haemophilus influenzae and Moraxella catarrhalis. Antimi-
impaired hepatic vitamin K metabolism in patients treated
crob Agents Chemother. 1982;22:859-877. crob Agents Chemother. 2006;50:2050-2057.
with N-methyl-thiotetrazole cephalosporins. Thromb
40. Jones RN. Review of the in-vitro spectrum and character- 61. Amsler KM, Davies TA, Shang W, et al. In vitro activity of
Haemost. 1984;51:358-361.
istics of cefmetazole (CS-1170). J Antimicrob Chemother. ceftibiprole against pathogens from two phase 3 clinical
13. Buening MK, Wold JS, Israel KS, et al. Disulfiram-like reac-
1989;12(suppl D):1-12. trials of complicated skin and skin structure infections.
tions to β-lactams. JAMA. 1981;245:2027.
41. Goldstein EJC, Citron DM. Annual incidence, epidemiol- Antimicrob Agents Chemother. 2008;52:3418-3423.
14. Hanaki H, Akagi H, Nomura S, et al. Structure-activity
ogy and comparative in vitro susceptibilities to cefoxitin, 62. Brown SD, Traczewski MM. Comparative in vitro antimi-
relationships of cephalosporin derivatives against
cefotetan, cefmetazole and ceftizoxime of recent crobial activity of a new cephalosporin, ceftaroline, and
methicillin-resistant Staphylococcus aureus and Enterococ-
community-acquired isolates of the Bacteroides fragilis determination of quality control ranges for MIC testing.
cus faecalis. J Antibiot. 1996;49:402-404.
group. J Antimicrob Chemother. 1988;26:2361-2366. Antimicrob Agents Chemother. 2009;53:1271-1274.
15. Davies TA, Page MGP, Shang W, et al. Binding of ceftobi-
42. Barry AL, Jones RN. Cefixime: spectrum of antibacterial 63. Ge Y, Biek D, Talbot GH, et al. In vitro profiling of ceftaro-
prole and comparators to the penicillin-binding proteins of
activity against 16016 clinical isolates. Pediatr Infect Dis J. line against a collection of recent bacterial clinical isolates
Escherichia coli, Pseudomonas aeruginosa, Staphylococcus
1987;6:954-957. from across the United States. Antimicrob Agents Che-
aureus, and Streptococcus pneumoniae. Antimicrob Agents
43. King A, Boothman C, Phillips I. Comparative in vitro mother. 2008;52:3398-3407.
Chemother. 2007;51:2621-2624.
activity of cefpirome and cefepime, two new cephalospo- 64. Chow AW, Finegold SM. In-vitro activity of ceftizoxime
16. Ikeda Y, Ban J, Ishikawa T, et al. Stability and stabilizing
rins. Eur J Clin Microbiol Infect Dis. 1990;9:677-685. against anaerobic bacteria and comparison with other
studies of TAK-599 (ceftaroline fosamil), a novel
44. Arguedas AG, Arrieta AC, Sutman HR, et al. In-vitro activ- cephalosporins. J Antimicrob Chemother. 1982;10(suppl C):
N-phosphono type prodrug of anti-methicillin resistant
ity of cefprozil (BMY 28100) and loracarbef (LY 163892) 45-50.
Staphylococcus aureus cephalosporin T-91825. Chem
against pathogens obtained from middle ear fluid. J Anti- 65. Murray RP, Jones RN, Allen SD, et al. Multilaboratory
Pharm Bull (Tokyo). 2008;56:1406-1411.
microb Chemother. 1991;27:311-318. evaluation of the in vitro activity of 13 β-lactam antibiotics
17. Schmidt-Hoffman A, Roos B, Schleimer M, et al. Single
45. Bragman SGL, Casewell MW. The in-vitro activity of ceft- against 1474 clinical isolates of aerobic and anaerobic bac-
dose pharmacokinetics and safety of a novel broad-
ibuten against 475 clinical isolates of gram-negative bacilli, teria. Diagn Microbiol Infect Dis. 1993;16:191-203.
spectrum cephalosporin (BAL 5788) in healthy volunteers.
compared with cefuroxime and cefadroxil. J Antimicrob 66. Ferrari M, Galla F, Pagnes P. In vitro antitreponemic activ-
Antimicrob Agents Chemother. 2004;48:2570-2575.
Chemother. 1990;25:221-226. ity of twelve cephalosporins. Chemotherapy. 1965;10:
18. Shlaes DM. New β-lactam-β-lactamase inhibitor combina-
46. Jones RN, Barry AL, Collaborative Antimicrobial Suscep- 305-311.
tions in clinical development. Ann N Y Acad Sci. 2013;
tibility Testing Group. Ceftibuten (7432-S, SCH 39720): 67. Hunfeld KP, Rodel R, Wichelhaus TA. In vitro activity of
1277:105-114.
comparative antimicrobial activity against 4735 clinical eight oral cephalosporins against Borrelia burgdorferi. Int J
19. Wise EM Jr, Park JT. Penicillin: its basic site of action as
isolates, beta-lactamase stability and broth microdilution Antimicrob Agents. 2003;21:313-318.
an inhibitor of a peptide cross-linking reaction in cell wall
quality control guidelines. Eur J Clin Microbiol Infect Dis. 68. Segreti J, Kapell KS, Trenholme GM. In vitro activity of
mucopeptide synthesis. Proc Natl Acad Sci U S A. 1965;
1988;7:802-807. beta-lactam drugs and sulbactam against Chlamydia tra-
54:75-81.
47. Debbia EA, Pesce A, Chiesa M, et al. Microbiologic profile chomatis. Diagn Microbiol Infect Dis. 1992;15:371-373.
20. Virudachalam R, Rao VS. Theoretical studies on beta-
of ceftibuten, a new oral cephalosporin. Drugs Exp Clin Res. 69. Cormican MG, Jones RN. Antimicrobial activity of cefo-
lactam antibiotics: I. Conformational similarity of penicil-
1992;18:129-139. taxime tested against infrequently isolated pathogenic
lin and cephalosporins to X-d-alanyl-d-alanine and
48. Briggs BN, Jones RN, Erwin ME, et al. In vitro activity species (unusual pathogens). Diagn Microbiol Infect Dis.
correlation of their structure with activity. Int J Peptide
evaluations of cefdinir (FK482, Cl-983, and PD134393): a 1995;22:43-48.
Protein Res. 1977;10:51-59.
novel orally administered cephalosporin. Diagn Microbiol 70. Stewart SM, Burnet ME, Young JE. In vitro sensitivity of
21. Waxman DJ, Strominger JL. Penicillin binding proteins and
Infect Dis. 1991;14:425-434. strains of mycoplasmas from human sources to antibiotics
the mechanism of action of beta-lactam antibiotics. Annu
49. Jones RN, Biedenbach DJ, Croco MAT, et al. In vitro and to sodium aurothiomalate and tylosin tartrate. J Med
Rev Biochem. 1983;52:825-869.
evaluation of a novel orally administered cephalosporin Microbiol. 1969;2:287-292.
22. Spratt BG. Distinct penicillin-binding protein involved in
(cefditoren) tested against 1249 recent clinical isolates of 71. Livermore DM. Mechanisms of resistance to cephalosporin
the division, elongation and shape of Escherichia coli K12.
Haemophilus influenzae, Moraxella catarrhalis, and Strep- antibiotics. Drugs. 1987;34(suppl 2):64-88.
Proc Natl Acad Sci U S A. 1975;72:2999-3003.
tococcus pneumoniae. Diagn Microbiol Infect Dis. 1998;31: 72. Sanders CC. Cefepime. Clin Infect Dis. 1993;17:369-379.
23. Buijs J, Dofferhoff AS, Mouton JW, et al. Concentration-
573-578. 73. Choi SH, Lee JE, Park SJ, et al. Emergence of antibiotic
dependency of beta-lactam-induced filament formation
50. Clark CE, Naomi K, Dewasse BE, et al. Activity of cefdito- resistance during therapy for infections caused by Entero-
in gram-negative bacteria. Clin Microbiol Infect. 2008;14:
ren against respiratory pathogens. Int J Antimicrob Agents. bacteriaceae producing AmpC beta-lactamases. Antimi-
344-349.
2002;50:33-41. crob Agents Chemother. 2008;52:995-1000.
24. Vogelman B, Craig WA. Kinetics of antimicrobial activity.
51. Jones RN, Kirby JT, Rhomberg PR. Comparative activity of 74. Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev.
J Pediatr. 1986;108:835-840.
meripenem in US medical centers (2007): initiating the 2009;22:161-182.
25. Bundtzen RW, Gerber AU, Cohn D, et al. Postantibiotic
2nd decade of MYSTIC program surveillance. Diagn 75. Sanders WE Jr, Sanders CC. Inducible β-lactamases: clini-
suppression of bacterial growth. Rev Infect Dis. 1981;3:
Microbiol Infect Dis. 2008;61:203-213. cal and epidemiologic implications for use of newer cepha-
28-37.
52. Karlowsky JA, Jones MY, Hayfield DC, et al. Ceftriaxone losporins. Rev Infect Dis. 1988;10:830-838.
26. Craig WA. Interrelationship between pharmacokinetics
activity against gram-positive and gram-negative patho- 76. Medeiros AA. Evolution and dissemination of β-lactamases
and pharmacodynamics in determining dosage regimens
gens isolated in US clinical microbiology laboratories from accelerated by generations of β-lactam antibiotics. Clin
for broad-spectrum cephalosporins. Diagn Microbiol Infect
1996 to 2000: results from The Surveillance Network (TSN) Infect Dis. 1997;24(suppl 1):Sl9-S45.
Dis. 1995;22:89-96.
Database-USA. Int J Antimicrob Agents. 2002;19:413-426. 77. Paterson DL, Bonomo RA. Extended-spectrum β-
27. Craig WA. Pharmacokinetic/pharmacodynamic parame-
53. Pfaller MA, Sader HS, Fritsche TR, et al. Antimicrobial lactamases: a clinical update. Clin Microbiol Rev. 2005;18:
ters: rationale for antibacterial dosing of mice and men.
activity of cefepime tested against ceftazidime-resistant 657-686.
Clin Infect Dis. 1998;26:1-12.
gram-negative clinical strains from North American Hos- 78. Bush K. Proliferation and significance of clinically relevant
28. Craig WA. Basic pharmacodynamics of antibacterials
pitals: report from the SENTRY Antimicrobial Surveillance β-lactamases. Ann N Y Acad Sci. 2013;1277:84-90.
with clinical applications to the use of beta-lactams,
292.e2
79. Quennan AM, Shang W, Kania M, et al. Interaction of 102. Nikaido H. Multidrug efflux pumps of gram-negative bac- 129. Craig WA, Suh B. Protein binding and the antimicrobial
ceftibiprole with beta-lactamases from molecular classes A teria. J Bacteriol. 1996;178:5853-5859. effects: methods for the determination of protein binding.
to D. Antimicrob Agents Chemother. 2007;51:3089-3095. 103. Nagai K, Davies TA, Jacobs MR, et al. Effects of amino In: Lorian V, ed. Antibiotics in Laboratory Medicine. Balti-
80. Mushtaq S, Warner M, Ge Y, et al. In vitro activity of cef- acid alterations in penicillin-binding proteins (PBPs) 1a, more: Williams & Wilkins; 1991:367-402.
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

taroline (PPI-0903M, T-91825) against bacteria with 2b, and 2x on PBP affinities of penicillin, ampicillin, amoxi- 130. Friedrich H, Haensel-Friedrich G, Langmaak H, et al.
defined resistance mechanisms and phenotypes. J Antimi- cillin, cefditoren, cefuroxime, cefprozil, and cefaclor in 18 Investigations of cefuroxime levels in the cerebrospinal
crob Chemother. 2007;60:300-311. clinical isolates of penicillin-susceptible, -intermediate, fluid of patients with and without meningitis. Chemother-
81. Paterson DL, Hujer KM, Hujer AM, et al, International and -resistant pneumococci. Antimicrob Agents Chemother. apy. 1980;26:91-97.
Klebsiella Study Group. Extended-spectrum beta- 2002;46:1273-1280. 131. Cable D, Edralin G, Overturf GD. Human cerebrospinal
lactamases in Klebsiella pneumoniae bloodstream isolates 104. du Plessis M, Smith AM, Klugman KP. Analysis fluid pharmacokinetics and treatment of bacterial menin-
from seven countries: dominance and widespread preva- of penicillin-binding protein 1b and 2a genes from gitis with ceftizoxime. J Antimicrob Chemother. 1982;
lence of SHV- and CTX-M-type beta-lactamases. Antimi- Streptococcus pneumoniae. Microb Drug Resist. 2000;6: 10(suppl C):121-127.
crob Agents Chemother. 2003;47:3554-3560. 127-131. 132. Chandrasekar PH, Rolston KV, Smith BR, et al. Diffusion
82. D’Andrea MM, Arena F, Pallecchi L, Rossolini GM. CTX- 105. Banerjee R, Gretes M, Basuino L, et al. In vitro selection of ceftriaxone into the cerebrospinal fluid of adults. J Anti-
M-type β-lactamases: a successful story of antibiotic resis- and characterization of ceftobiprole-resistant methicillin- microb Chemother. 1984;14:427-430.
tance. Int J Med Microbiol. 2013;303:305-317. resistant Staphylococcus aureus. Antimicrob Agents Che- 133. Cherubin CE, Eng RHK, Norrby R, et al. Penetration of
83. Pallett A, Hand K. Complicated urinary tract infections: mother. 2008;52:2089-2096. newer cephalosporins into cerebrospinal fluid. Rev Infect
practical solutions for the treatment of multiresistant 106. Barza M, Melethil S, Berger S, et al. Comparative pharma- Dis. 1989;11:526-548.
gram-negative bacteria. J Antimicrob Chemother. 2010; cokinetics of cefamandole, cephapirin, and cephalothin in 134. Fong IW, Tomkins KB. Penetration of ceftazidime into the
65(suppl 3):25-33. healthy subjects and effect of repeated dosing. Antimicrob cerebrospinal fluid of patients with and without evidence
84. Queenan AM, Bush K. Carbapenemases: the versatile Agents Chemother. 1976;10:421-425. of meningeal inflammation. Antimicrob Agents Chemother.
β-lactamases. Clin Microbiol Rev. 2007;20:440-458. 107. Pfeffer M, Jackson A, Ximenes J, et al. Comparative human 1984;26:115-116.
85. Pfaller MA, Jones RN. Antimicrobial susceptibility of oral clinical pharmacology of cefadroxil, cephalexin, and 135. Wolff M, Chavanet P, Kazmierczak A, et al. Diffusion of
inducible AmpC beta-lactamase-producing Enterobacte- cephradine. Antimicrob Agents Chemother. 1977;11: cefpirome into the cerebrospinal fluid of patients with
riaceae from the Meropenem Yearly Susceptibility Test 331-338. purulent meningitis. J Antimicrob Chemother. 1992;29
Information Collection (MYSTIC) programme, Europe 108. Korzeniowski OM, Scheld WM, Sande MA. Comparative (suppl A):59-62.
1997-2000. Int J Antimicrob Agents. 2002;19:383-388. pharmacology of cefaclor and cephalexin. Antimicrob 136. Andes D, Craig WA. Pharmacokinetics and pharmaco­
86. Endimiani A, Perez F, Bonomo RA. Cefepime: a reappraisal Agents Chemother. 1977;12:157-162. dynamics of outpatient intravenous antimicrobial therapy.
in an era of increasing antimicrobial resistance. Expert Rev 109. Patel IH, Chen S, Parsonnet M, et al. Pharmacokinetics of Infect Dis Clinics North Am. 1998;12:849-859.
Anti Infect Ther. 2008;6:805-824. ceftriaxone in humans. Antimicrob Agents Chemother. 137. Strehl E, Kees F. Pharmacological properties of parenteral
87. Clinical Laboratory and Standards Institute. Performance 1981;20:634-640. cephalosporins: rationale for ambulatory use. Drugs. 2000;
Standards for Antimicrobial Susceptibility Testing; Twenty- 110. Brogard JM, Conte E. Pharmacokinetics of the new cepha- 59(suppl 3):9-18.
first Informational Supplement. Document M100-S21 (M2 losporins. Antibiot Chemother. 1982;31:145-210. 138. Johnson CA, Zimmerman SW, Rogge M. The pharmacoki-
and M7). Wayne, PA: Clinical Laboratory and Standards 111. Cutler RE, Blair AD, Burgess ED. Pharmacokinetics of netics of antibiotics used to treat peritoneal dialysis-
Institute; 2011. ceftizoxime. J Antimicrob Chemother. 1982;10(suppl C): associated peritonitis. Am J Kidney Dis. 1984;4:3-17.
88. Poirel L, Naas T, Le Thomas I, et al. CTX-M-Type 91-97. 139. Groneberg DA, Doring F, Eynott PR, et al. Intestinal
extended-spectrum β-lactamase that hydrolyzes ceftazi- 112. Barriere SL, Flaherty JR. Third generation cephalosporins: peptide transport: ex vivo uptake studies and localization
dime through a single amino acid substitution in the a critical evaluation. Clin Pharm. 1984;3:351-373. of peptide carrier PEPT1. Am J Physiol Gastrointest Liver
omega loop. Antimicrob Agents Chemother. 2001;45: 113. Browning MJ, Holt HA, White LO, et al. Pharmacokinetics Physiol. 2001;281:697-704.
3355-3361. of cefotetan in patients with end-stage renal failure on 140. Saab AN, Dittert LW, Hussain AA. Isomerization of cepha-
89. Lewis JS 2nd, Herrera M, Wickes B, et al. First report of the maintenance dialysis. J Antimicrob Chemother. 1986;18: losporin esters: implications for the prodrug ester approach
emergence of CTX-M-type extended-spectrum beta- 103-106. to enhancing the oral bioavailabilities of cephalosporins.
lactamases (ESBLs) as the predominant ESBL isolated in a 114. Faulkner RD, Bohaychuk W, Haynes JD, et al. The pharma- J Pharm Sci. 1988;77:906-907.
U.S. health care system. Antimicrob Agents Chemother. cokinetics of cefixime in the fasted and fed state. Eur J Clin 141. Redington J, Ebert SC, Craig WA. Role of antimicrobial
2007;51:4015-4021. Pharmacol. 1988;34:525-528. pharmacokinetics and pharmacodynamics in surgical pro-
90. Castanheira M, Mendes RE, Rhomberg PR, et al. Rapid 115. Barbhaiya RH, Shukla UA, Gleason CR, et al. Compari- phylaxis. Rev Infect Dis. 1991;13(suppl 10):S790-S799.
emergence of blaCTX-M among Enterobacteriaceae in US son of the effects of food on the pharmacokinetics of cef- 142. Joukhadar C, Derendorf H, Muller M. Microdialysis: a
Medical Centers: molecular evaluation from the MYSTIC prozil and cefaclor. Anticrob Agents Chemother. 1990;34: novel tool for clinical studies of anti-infective agents. Eur J
Program (2007). Microb Drug Resist. 2008;14:211-216. 1210-1213. Clin Pharmacol. 2001;57:211-219.
91. Williamson DA, Roberts SA, Smith M, et al. High rates of 116. Wise R. The pharmacokinetics of the oral cephalosporins: 143. Barbour A, Schmidt S, Sabarinath SN, et al. Soft-tissue
susceptibility to ceftazidime among globally prevalent a review. J Antimicrob Chemother. 1990;26(suppl E): penetration of ceftopibrole in healthy volunteers deter-
CTX-M-producing Escherichia coli: potential clinical 13-20. mined by in-vivo microdialysis. Antimicrob Agents Che-
implications of the revised CLSI interpretive criteria. Eur J 117. Barr WH, Lin CC, Radwanski E, et al. The pharmacokinet- mother. 2009;53:2773-2776.
Clin Microbiol Infect Dis. 2012;31:821-824. ics of ceftibuten in humans. Diagn Microbiol Infect Dis. 144. Higa F, Kusano N, Tateyama M, et al. Simplified quantita-
92. Kahlmeter G. Breakpoints for intravenously used cephalo- 1991;14:93-100. tive assay system for measuring activities of drugs against
sporins in Enterobacteriaceae: EUCAST and CLSI break- 118. Barbhaiya RH, Forgue ST, Gleason CR. Pharmacokinetics intracellular Legionella pneumophila. J Clin Microbiol.
points. Clin Microbiol Infect. 2008;14(suppl 1):169-174. of cefepime after single and multiple intravenous adminis- 1998;36:1392-1398.
93. Dudley MN, Ambrose PG, Bhavnani SM, et al. Background trations in healthy subjects. Antimicrob Agents Chemother. 145. Andes D, Craig WA. Pharmacokinetics and pharmacody-
and rationale for revised Clinical and Laboratory Standards 1992;36:552-557. namics of antibiotics in meningitis. Infect Dis Clin North
Institute interpretive criteria (breakpoints) for Enterobac- 119. DeSante KA, Zeckel ML. Pharmacokinetic profile of lor- Am. 1999;13:595-617.
teriaceae and Pseudomonas aeruginosa: 1. Cephalosporins acarbef. Am J Med. 1992;92(suppl 6A):16S-25S. 146. Dacey RG, Sande MA. Effect of probenecid on cerebrospi-
and aztreonam. Clin Infect Dis. 2013;56:1301-1309. 120. Craig WA. The pharmacokinetics of cefpirome: rationale nal fluid concentrations of penicillin and cephalosporin
94. Livermore DM, Andrews JM, Hawkey PM, et al. Are sus- for a twelve-hour dosing regimen. Scand J Infect Dis Suppl. derivatives. Antimicrob Agents Chemother. 1974;6:437-441.
ceptibility tests enough, or should laboratories still seek 1993;91:33-40. 147. Jones RN. A review of cephalosporin metabolism: a lesson
ESBLs and carbapenemases directly? J Antimicrob Che- 121. Nix DE, Symonds WT, Hyatt JM, et al. Comparative phar- to be learned for future chemotherapy. Diagn Microbiol
mother. 2012;67:1569-1577. macokinetics of oral ceftibuten, cefixime, cefaclor, and Infect Dis. 1989;12:25-31.
95. Lee NY, Lee CC, Huang WH, et al. Cefepime therapy for cefuroxime axetil in healthy volunteers. Pharmacotherapy. 148. Chin NX, Neu HC. Cefotaxime and desacetyl cefotaxime:
monomicrobial bacteremia caused by cefepime-susceptible 1997;17:121-125. an example of advantageous antimicrobial metabolism.
extended-spectrum beta-lactamase-producing Enterobac- 122. Li JT, Hou F, Lu H. Phase I clinical trial of cefditoren pivoxil Diagn Microbial Infect Dis. 1984;2(suppl 3):21S-31S.
teriaceae: MIC matters. Clin Infect Dis. 2013;56:488-495. (ME 1207): pharmacokinetics in healthy volunteers. Drugs 149. Craig WA. Pharmacokinetics of antibiotics with special
96. Martinez-Martinez L, Hernandez-Alles S, Alberti S, et al. Under Exp Clin Res. 1997;23:145-150. emphasis on cephalosporins. Clin Microbiol Infect. 2000;
In vivo selection of porin-deficient mutants of Klebsiella 123. Guay DR. Cefdinir: an advanced-generation, broad- 6(suppl 3):46-49.
pneumoniae with increased resistance to cefoxitin and spectrum oral cephalosporin. Clin Ther. 2002;24:473-489. 150. Craig WA, Vogelman B. Changing patterns of hospital
expanded-spectrum-cephalosporins. Antimicrob Agents 124. Talbot GH, Thye D, Das A, et al. Phase 2 study of ceftaroline infections: implications for therapy. Changing concepts
Chemother. 1996;40:342-348. versus standard therapy in treatment of complicated skin and new applications of antibiotic pharmacokinetics. Am J
97. Charrel RN, Pages JM, De Micco P, et al. Prevalence of and skin structure infections. Antimicrob Agents Che- Med. 1984;77(suppl 1B):24-28.
outer membrane porin alteration in beta-lactam-antibiotic- mother. 2007;51:3612-3616. 151. Aronoff GR, Bennett WM, Berns JS, et al. Drug Prescribing
resistant Enterobacter aerogenes. Antimicrob Agents Che- 125. Schmitt-Hoffmann A, Nyman L, Roos B, et al. Multiple- in Renal Failure: Dosing Guidelines for Adults. 4th ed. Phila-
mother. 1996;40:2854-2858. dose pharmacokinetics and safety of a novel broad- delphia: American College of Physicians–American
98. Mazzariol A, Cornaglia G, Nikaido H. Contributions of the spectrum cephalosporin (BAL5788) in healthy volunteers. Society of Internal Medicine; 1999.
AmpC β-lactamase and the AcrAB multidrug efflux system Antimicrob Agents Chemother. 2004;48:2576-2580. 152. Livornese LL Jr, Slavin D, Benz RL, et al. Use of antibacte-
in intrinsic resistance of Escherichia coli K-12 to β-lactams. 126. Kimko HH, Murthy B, Xu XS, et al. Population pharmaco- rial agents in renal failure. Infect Dis Clin North Am. 2000;
Antimicrob Agents Chemother. 2000;44:1387-1390. kinetic analysis of ceftibiprole for the treatment of compli- 14:371-390.
99. Masuda N, Sakagawa E, Ohya S, et al. Substrate specificities cated skin and skin-structure infections. Antimicrob Agents 153. Bressolle F, Kinowski JM, de la Coussaye JE, et al. Clinical
of MexAB-OprM, MexCD-OprJ, and MexXY-OprM efflux Chemother. 2009;53:1228-1230. pharmacokinetics during continuous haemofiltration. Clin
pumps in Pseudomonas aeruginosa. Antimicrob Agents 127. Ge Y, Whitehouse MJ, Friedland I, Talbot GH. Pharmaco- Pharmacokinet. 1994;26:457-471.
Chemother. 2000;44:3322-3327. kinetics and safety of CXA-101, a new antipseudomonal 154. Baumgart KW, Baldo BA. Cephalosporin allergy. N Engl J
100. Hocquet D, Nordman P, El Garch F, et al. Involvement of cephalosporin, in healthy adult male and female subjects Med. 2002;346:380-381.
the MexXY-OprM efflux system in emergence of cefepime receiving single- and multiple-dose intravenous infusions. 155. Macy E. Cephalosporin allergy. N Engl J Med. 2002;346:
resistance in clinical strains of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2010;54:3427-3431. 380-381.
Antimicrob Agents Chemother. 2006;50:1347-1351. 128. Miller B, Hershberger E, Benziger D, et al. Pharmacokinet- 156. Norrby SR. Side effects of cephalosporins. Drugs. 1987;
101. Baum EZ, Crespo-Carbone SM, Morrow BJ, et al. Effect of ics and safety of intravenous ceftolozane-tazobactam in 34(suppl 2):105-120.
MexXY overexpression on ceftobiprole susceptibility in healthy adult subjects following single and multiple 157. Romano A, Mayorga C, Torres MJ, et al. Immediate allergic
Pseudomonas aeruginosa. Antimicrob Agents Chemother. ascending doses. Antimicrob Agents Chemother. 2012;56: reaction to cephalosporins: cross-reactivity and selective
2009;53:2785-2790. 3086-3091. responses. J Allergy Clin Immunol. 2000;106:1177-1183.
292.e3
158. Hebert AA, Sigman ES, Levy ML. Serum sickness-like 188. Inagaki J, Bodey GP. Phlebitis associated with cephalospo- 216. McCarty JM, Renteria A. Treatment of pharyngitis and
reactions from cefaclor in children. J Am Acad Dermatol. rins: cephapirin versus cephalothin. Curr Ther Res Clin tonsillitis with cefprozil: review of three multicenter trials.
1991;25:805-808. Exp. 1973;15:37-43. Clin Infect Dis. 1992;14(suppl 2):S224-S230.
159. Pichichero ME. A review of evidence supporting the 189. Thompson JW, Jacobs RF. Adverse effects of newer cepha- 217. Nadelman RB, Luger SW, Frank E, et al. Comparison of

Chapter 21  Cephalosporins


American Academy of Pediatrics recommendation for pre- losporins: an update. Drug Saf. 1993;9:132-142. cefuroxime axetil and doxycycline in the treatment of early
scribing cephalosporin antibiotics for penicillin-allergic 190. Briggs GG, Freeman RK, Yaffee SJ. Drugs in Pregnancy and Lyme disease. Ann Intern Med. 1992;117:273-280.
patients. Pediatrics. 2005;115:1048-1057. Lactation: A Reference Guide to Fetal and Neonatal Risk. 218. Poindexter AN, Sweet R, Ritter M. Cefotetan in the treat-
160. Miranda A, Blanca M, Vega JM, et al. Cross-reactivity 2nd ed. Baltimore: Williams & Wilkins; 1986. ment of obstetric and gynecologic infections. Am J Obstet
between a penicillin and a cephalosporin with the same 191. McManus MC, Barriere SL. Interaction between newer Gynecol. 1986;154:946-950.
side chain. J Allergy Clin Immunol. 1996;98:671-677. cephalosporins and Clinitest, Diastix, and Tes-Tape. Am J 219. Wilson SE, Boswick JA, Duma RJ, et al. Cephalosporin
161. Camagna JD, Bond MC, Schabelman E, et al. The use of Hosp Pharm. 1983;40:1544-1545. therapy in intraabdominal infections: a multicenter ran-
cephalosporins in penicillin-allergic patients: a literature 192. Letellier G, Desjarlais F. Analytical interference of drugs in domized, comparative study of cefotetan, moxalactam and
review. J Emerg Med. 2012;42:612-620. clinical chemistry: II. The interference of three cephalospo- cefoxitin. Am J Surg. 1988;155(suppl 5A):61-66.
162. Pichichero ME, Casey JR. Safe use of selected cephalospo- rins with the determination of serum creatinine concentra- 220. Sweet R, Gall SA, Gobbs RS, et al. Multicenter clinical trial
rins in penicillin-allergic patients: a meta-analysis. Otolar- tion by the Jaffe reaction. Clin Biochem. 1985;18:352-356. comparing cefotetan with moxalactam or cefoxitin as
yngol Head Neck Surg. 2007;136:340-347. 193. Thomakos N, Maraki S, Liakakos T, et al. Effect of cefaman- therapy for obstetric gynecologic infections. Am J Surg.
163. Nugent JS, Quinn JM, McGrath CM, et al. Determination dole, cefuroxime and cefoxitin on yeast fecal flora of surgi- 1988;155(suppl 5A):S56-S60.
of the incidence of sensitization after penicillin skin testing. cal patients. Chemotherapy. 1998;44:324-327. 221. Appelman MD, Haseltine PNR, Cherubin CE. Epidemiol-
Ann Allergy Asthma Immunol. 2003;90:398-403. 194. MacDonald TM, Beardon PH, McGilchrist MM, et al. The ogy, antimicrobial susceptibility, pathogenicity, and signifi-
164. Smith CR. Considerations regarding clinical safety and risks of symptomatic vaginal candidiasis after oral antibi- cance of Bacteroides fragilis group organisms isolated at Los
tolerability of antibiotics in serious and nosocomial infec- otic therapy. J Med. 1993;86:419-424. Angeles County–University of Southern California
tions. Clin Ther. 1981;4(suppl A):133-145. 195. Bartlett JG, Willey SH, Chang TW, et al. Cephalosporin- Medical Center. Rev Infect Dis. 1991;13:12-18.
165. Bang N, Kammer RB. Hematological complications asso­ associated pseudomembranous colitis due to Clostridium 222. Morel C, Vergnaud M, Lengeard MM, et al. Cefotetan:
ciated with beta-lactam antibiotics. Rev Infect Dis. 1983; difficile. JAMA. 1979;242:2683-2685. comparative study in vitro against 266 gram-negative clini-
5(suppl 2):S380-S393. 196. Weiss B, Kleinkauf N, Eckmanns T, et al. Risk factors cal isolates. J Antimicrob Chemother. 1983;11(suppl A):
166. Garratty G. Immune hemolytic anemia caused by drug. related to a hospital-associated cluster of Clostridium dif- 31-36.
Expert Opin Drug Saf. 2012;11:635-642. ficile PCR ribotype 027 infections in Germany during 2007. 223. Snydman DR, Jacobus NV, McDermott LP, et al. National
167. Wong BB, Ko GJ. Neutropenia in patients receiving long- Infect Control Hosp Epidemiol. 2009;30:282-284. survey of the susceptibility of Bacteroides fragilis group:
term cefepime therapy for osteomyelitis. Am J Health Syst 197. Grayson ML, McDonald M, Gibson K, et al. Once-daily report and analysis of trends in the United States from 1997
Pharm. 2003;60:2229-2232. intravenous cefazolin plus oral probenecid is equivalent to to 2004. Antimicrob Agents Chemother. 2007;51:1649-1655.
168. Molthan L, Reidenberg MM, Eichman MF. Positive direct once-daily intravenous ceftriaxone plus oral placebo for the 224. Centers for Disease Control and Prevention. Sexually
Coombs tests due to cephalothin. N Engl J Med. 1967;277: treatment of moderate-to-severe cellulitis in adults. Clin transmitted diseases treatment guidelines, 2010. MMWR
123-125. Infect Dis. 2002;34:1440-1448. Morb Mortal Wkly Rep. 2010;51(RR-12):1-110.
169. Ingalls CS, Freimer EH. Detection of antibiotic-induced 198. Regamey C, Lobke RD, Engelking ER, et al. Inactivation of 225. Unemo M, Nicholas RA. Emergence of multidrug-
platelet dysfunction in whole blood using flow cytometry. cefazolin, cephaloridine, and cephalothin by methicillin- resistant, extensively drug-resistant and untreatable gonor-
J Antimicrob Chemother. 1992;29:313-321. sensitive and methicillin-resistant strains of Staphylococcus rhoea. Future Microbiol. 2012;7:1401-1422.
170. Barza M, Furie B, Brown AK, et al. Defects in vitamin aureus. J Infect Dis. 1975;131:291-294. 226. Peterson HB, Galaid EI, Zenilman JM. Pelvic inflammatory
K-dependent carboxylation associated with moxalactam 199. Fong IW, Engelking ER, Kirby WMM. Relative inactivation disease: review of treatment options. Rev Infect Dis.
treatment. J Infect Dis. 1986;153:1166-1169. by Staphylococcus aureus of eight cephalosporin antibiotics. 1990;12(suppl 6):S656-S664.
171. Shearer MJ, Bechtold H, Andrassy K, et al. Mechanism of Antimicrob Agents Chemother. 1976;9:939-944. 227. Trenholme GM, Schmitt BA, Nelson JA, et al. Comparative
cephalosporin-induced hypoprothrombinemia: relation to 200. Bratzler DW, Dellinger EP, Olsen KM, et al. Clinical prac- study of three different dosing regimens of cefotaxime for
cephalosporin side chain, vitamin K metabolism, and tice guideline for antimicrobial prophylaxis in surgery. Am gram-negative bacteremia. Diagn Microbiol Infect Dis.
vitamin K status. J Clin Pharmacol. 1988;28:88-95. J Health-Syst Pharm. 2013;70:195-283. 1989;12:107-112.
172. Conly JM, Ramotar K, Chubb H, et al. Hypoprothrombin- 201. Weber DJ, Wolfson JS, Swartz MN, et al. Pasteurella mul- 228. Young JPW, Husson JM, Bruch K, et al. The evaluation of
emia in febrile, neutropenic patients with cancer: associa- tocida infections: report of 3 cases and review of the litera- efficacy and safety of cefotaxime: a review of 2500 cases.
tion with antimicrobial suppression of intestinal microflora. ture. Medicine (Baltimore). 1984;63:133-154. J Antimicrob Chemother. 1980;6(suppl A):293-300.
J Infect Dis. 1984;150:202-212. 202. Milatovic D. Evaluation of cefadroxil, penicillin and eryth- 229. Daikos GK, Kosmidis J, Giamarellou H, et al. Evaluation of
173. Baillargeon J, Holmes HM, Lin YL, et al. Concurrent use romycin in the treatment of streptococcal tonsillopharyn- cefotaxime in a hospital with high antibiotic resistance
of warfarin and antibiotics and the risk of bleeding in older gitis. Pediatr Infect Dis J. 1991;10(suppl 10):S61-S63. rates. J Antimicrob Chemother. 1980;6(suppl A):255-261.
adults. Amer J Med. 2012;125:183-189. 203. Disney FA, Dillon H, Blumer JL, et al. Cephalexin and 230. Eron LJ, Park CH, Goldenberg RI, et al. Ceftriaxone
174. Fainstein V, Bodey GP, McCredie KB, et al. Coagulation penicillin in the treatment of group A β-hemolytic strep- therapy of serious bacterial infections. J Antimicrob Che-
abnormalities induced by beta-lactam antibiotics in cancer tococcal throat infections. Am J Dis Child. 1992;146: mother. 1983;12:65-78.
patients. J Infect Dis. 1983;148:745-750. 1324-1327. 231. Osmon DR, Berbari EF, Berendt AR, et al. Diagnosis and
175. Nichols RL, Wikler MA, McDevitt JT, et al. Coagulopathy 204. Norby SR. Short-term treatment of uncomplicated lower management of prosthetic joint infection: clinical practice
associated with extended-spectrum cephalosporins in urinary tract infections in women. Rev Infect Dis. 1990;12: guidelines by the Infectious Diseases Society of America.
patients with serious infections. Antimicrob Agents Che- 458-467. Clin Infect Dis. 2013;56:e1-e25.
mother. 1987;31:281-285. 205. Mehtar S, Parr JH, Morgan DJR. A comparison of cefurox- 232. Chow JW, Fine MJ, Shlaes DM, et al. Enterobacter bactere-
176. McCloskey RV. Spontaneous reports of bleeding: compari- ime and cotrimoxazole in severe respiratory tract infec- mia: clinical features and emergence of antibiotic resistance
son of N-methylthiotetrazole side chain (MTT) and tions. J Antimicrob Chemother. 1982;9:479-484. during therapy. Ann Intern Med. 1991;115:585-590.
non-MTT cephalosporins. J Infect Dis. 1988;158:1405. 206. Marks WA, Stutman HR, Marks MI, et al. Cefuroxime 233. Congeni BL, Bradley J, Hammerschlag MR. Safety and effi-
177. Aronoff GR, Wolen RL, Obermeyer BD, et al. Pharmaco- versus ampicillin plus chloramphenicol in childhood bac- cacy of once daily ceftriaxone for the treatment of bacterial
kinetics and protein binding of cefamandole and its terial meningitis: a multicenter randomized controlled meningitis. Pediatr Infect Dis. 1986;5:293-297.
1-methyl-1 H-tetrazole-5-thiol side chain in subjects with trial. J Pediatr. 1986;109:123-130. 234. Jacobs RJ, Wells TO, Steele RW, et al. A prospective ran-
normal and impaired renal function. J Infect Dis. 1986; 207. Schaad UB, Suter S, Gianella-Borradori A, et al. A com- domized comparison of cefotaxime vs. ampicillin and
153:1069-1074. parison of ceftriaxone and cefuroxime for the treatment of chloramphenicol for bacterial meningitis in children.
178. Foster RS, Raehl CL, Wilson HD. Disulfiram-like reactions bacterial meningitis in children. N Engl J Med. 1990;322: Pediatr Infect Dis. 1986;4:362-368.
associated with a parenteral cephalosporin. Am J Hosp 141-147. 235. Mullaney DT, John IF. Cefotaxime therapy: evaluation of its
Pharm. 1980;37:858-859. 208. Bartlett JG, Dowell SF, Mandell LA, et al. Practice guide- effects on bacterial meningitis, CSF drug levels, and bacte-
179. Thiim M, Freidman LS. Hepatotoxicity of antibiotics and lines for the management of community-acquired pneu- ricidal activity. Arch Intern Med. 1983;143:1705-1708.
antifungals. Clin Liver Dis. 2003;7:381-399. monia in adults. Clin Infect Dis. 2000;31:347-382. 236. del Rio M, McCracken GH Jr, Nelson JD, et al. Pharmaco-
180. Heim-Duthoy KL, Caperton EM, Pollock R, et al. Apparent 209. Vu VL, Chiou CCC, Feldman C, et al. An international kinetics and cerebrospinal fluid bactericidal activity of
biliary pseudolithiasis during ceftriaxone therapy. Antimi- prospective study of pneumococcal bacteremia: correlation ceftriaxone in the treatment of pediatric patients with bac-
crob Agents Chemother. 1990;34:1146-1149. with in vitro resistance, antibiotics administered, and clini- terial meningitis. Antimicrob Agents Chemother. 1982;22:
181. Park HZ, Lee SP, Schy A, et al. Ceftriaxone-associated gall- cal outcome. Clin Infect Dis. 2003;37:230-237. 622-627.
bladder sludge. Gastroenterology. 1991;100:1665-1670. 210. Mandell LA, Wunderink RG, Anzueto A, et al. Infectious 237. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guide-
182. Schaad UB, Wedgwood-Krucko J, Tschaeppeler H. Revers- Diseases Society of America/American Thoracic Society lines for management of bacterial meningitis. Clin Infect
ible ceftriaxone-associated biliary pseudo-lithiasis in chil- consensus guidelines on the management of community- Dis. 2004;39:1267-1284.
dren. Lancet. 1988;2:1411-1413. acquired pneumonia in adults. Clin Infect Dis. 2007;44 238. Pallares R, Capdevila Q, Linares J, et al. The effect of cepha-
183. Steadman E, Raisch DW, Bennett CL, et al. Evaluation of a (suppl 2):S27-S72. losporin resistance on mortality in adult patients with
potential clinical interaction between ceftriaxone and cal- 211. Barson WJ, Miller MA, Marcon MJ, et al. Cefuroxime nonmeningeal systemic pneumococcal infections. Am J
cium. Antimicrob Agents Chemother. 2010;54:1534-1540. therapy for bacteremic soft-tissue infections in children. Med. 2002;113:120-126.
184. Barza M. Nephrotoxicity of cephalosporins: an overview. Am J Dis Child. 1985;139:1141-1144. 239. Sahm DF, Thornsberry C, Mayfield DC, et al. In vitro
J Infect Dis. 1978;137(suppl):S60-S73. 212. Torkkeli T, Ruoppi P, Nuutinen J, Kari A. Changed clinical activities of broad-spectrum cephalosporins against non-
185. Grill MF, Maganti R. Cephalosporin-induced neurotoxic- course and current treatment of acute epiglottitis in adults meningeal isolates of Streptococcus pneumoniae: MIC
ity: clinical manifestations, potential pathogenic mecha- a 12-year experience. Laryngoscope. 1994;104:1503-1506. interpretation using NCCLS M100-S12 recommendations.
nisms, and the role of electroencephalographic monitoring. 213. Craig WA, Andes D. Pharmacokinetics and pharmacody- J Clin Microbiol. 2002;40:669-674.
Ann Pharmacol. 2008;42:1843-1850. namics of antibiotics in otitis media. Pediatr Infect Dis J. 240. Leibovitz E, Piglansky L, Raiz S, et al. Efficacy of a three-
186. Martinez-Rodriguez JE, Barriga FJ, Santamaria J, et al. 1996;15:255-259. day intramuscular ceftriaxone regimen in nonresponsive
Nonconvulsive status epilepticus associated with cephalo- 214. Dagan R, Klugman KP, Craig WA, et al. Evidence to acute otitis media. Pediatr Infect Dis J. 1998;17:1126-1131.
sporins in patients with renal failure. Am J Med. 2001; support the rationale that bacterial eradication in respira- 241. Leibovitz E, Piglansky L, Raiz S, et al. Bacteriologic and
111:115-119. tory tract infection is an important aim of antimicrobial clinical efficacy of one day vs. three day intramuscular cef-
187. Lamouth F, Buclin T, Pascual A, et al. High cefepime therapy. J Antimicrob Chemother. 2001;47:129-140. triaxone for treatment of nonresponsive acute otitis media
plasma concentrations and neurological toxicity in febrile 215. Bisno AL, Gerber MA, Gwaltney JM Jr, et al. Practice in children. Pediatr Infect Dis J. 2000;19:1040-1050.
neutropenic patients with mild impairment of renal func- guidelines for the diagnosis and management of group A 242. Blumer JL, McLinn SE, Deabate CA, et al. Multinational
tion. Antimicrob Agents Chemother. 2010;54:4360-4367. streptococcal pharyngitis. Clin Infect Dis. 2002;35:113-125. multicenter controlled trial comparing ceftibuten with
292.e4
cefaclor for the treatment of acute otitis media. Pediatr four weeks compared with ceftriaxone plus gentamicin patients with community-acquired pneumonia requiring
Infect Dis J. 1995;14:S115-S120. once daily for two weeks for treatment of endocarditis hospitalization. Int J Antimicrob Agents. 2012;39:240-246.
243. Harrison CJ, Chartrand SA, Pichichero ME. Microbiologic due to penicillin-susceptible streptococci. Clin Infect Dis. 292. File TM Jr, Low DE, Eckburg PB, et al. Integrated analysis
and clinical aspects of a trial of once daily cefixime com- 1998;27:1470-1474. of FOCUS 1 and FOCUS 2: randomized, double-blinded,
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

pared with twice daily cefaclor for treatment of acute otitis 267. Neu HC, Chin NX, Labthavikul P. The in vitro activity and multicenter phase 3 trials of the efficacy and safety of
media in infants and children. Pediatr Infect Dis J. 1993; beta lactamase stability of cefpirome (HR 810), a pyridine ceftaroline fosamil versus ceftriaxone in patients with
12:62-69. cephalosporin agent active against staphylococci, Entero- community-acquired pneumonia. Clin Infect Dis. 2010;
244. Mandel EM, Casselbrant ML, Kurs-Lasky M, et al. Efficacy bacteriaceae, and Pseudomonas aeruginosa. Infection. 51:1395-1405.
of ceftibuten compared with amoxicillin for otitis media 1985;13:146-155. 293. Ho TT, Cadena J, Childs LM, et al. Methicillin-resistant
with effusion in infants and children. Pediatr Infect Dis J. 268. King A, Boothman C, Phillips I. Comparative in vitro Staphylococcus aureus bacteremia and endocarditis treated
1996;15:409-414. activity of cefpirome and cefepime, two new cephalospo- with ceftaroline salvage therapy. J Antimicrob Chemother.
245. Block SL, Hedrick JA, Tyler RD. Comparative study of the rins. Eur J Clin Microbiol Infect Dis. 1990;9:677-685. 2012;67:1267-1270.
effectiveness of cefixime and penicillin V for the treatment 269. Garau J, Wilson W, Wood M, Carlet J. Fourth generation 294. Lin JC, Aung G, Thomas A, et al. The use of Ceftaroline
of streptococcal pharyngitis in children and adolescents. cephalosporins: a review of in vitro activity, pharmacoki- fosamil in methicillin-resistant Staphylococcus aureus
Pediatr Infect Dis J. 1992;11:919-925. netics, pharmacodynamics, and clinical utility. Clin Micro- endocarditis and deep-seated MRSA infections: a retro-
246. Pichichero ME, Gooch WM 3rd. Comparison of cefdinir biol Infect. 1997;3(suppl 1):S87-S101. spective case series of 10 patients. J Infect Chemother.
and penicillin V in the treatment of pediatric strepto­ 270. Jauregui L, Matzke D, Scott M, et al. Cefepime as treatment 2013;19:42-49.
coccal tonsillopharyngitis. Pediatr Infect Dis J. 2000;19: for osteomyelitis and other severe bacterial infections. 295. Werth BJ, Sakoulas G, Rose WE, et al. Ceftaroline increases
8171-8173. J Antimicrob Chemother. 1993;32(suppl B):141-149. membrane binding and enhances the activity of daptomy-
247. Pichichero ME, McLinn SE, Gooch WM III, et al. Ceftibu- 271. Arguedas AG, Stutman HR, Zaleska M, et al. Cefepime: cin against daptomycin-nonsusceptible vancomycin-
ten vs penicillin V in group A beta-hemolytic streptococ- pharmacokinetics and clinical response in patients with intermediate Staphylococcus aureus in a pharmacokinetic/
cal pharyngitis. Pediatr Infect Dis J. 1995;14(suppl 7): cystic fibrosis. Am J Dis Child. 1992;146:797-802. pharmacodynamic model. Antimicrob Agents Chemother.
S102-S107. 272. Tam VH, McKinnon PS, Akins RL, et al. Pharmacodynam- 2013;57:66-73.
248. Banfi A, Gavriele G, Hill-Juarez JM, et al, Ceftibuten ics of cefepime in patients with gram-negative infection. 296. Rose WE, Schultz LT, Andes D, et al. Addition of ceftaro-
Urinary Tract Infection International Study Group. Multi- J Antimicrob Chemother. 2002;50:425-428. line to daptomycin after emergence of daptomycin-
national comparative trial of ceftibuten and trimethoprim- 273. Hoepelman AI, Kieft H, Aoun M, et al. International com- nonsusceptible Staphylococcus aureus during therapy
sulfamethoxazole in the treatment of children with parative study of cefepime and ceftazidime in the treatment improves antibacterial activity. Antimicrob Agents Che-
complicated or recurrent urinary tract infections. Pediatr of serious bacterial infections. J Antimicrob Chemother. mother. 2012;56:5296-5302.
Infect Dis. 1993;12(suppl):S84-S91. 1993;32(suppl B):175-186. 297. Arias CA, Murray BE. Emergence and management of
249. Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical practice 274. Chandrasekar PH, Arnow PM. Cefepime versus ceftazi- drug-resistant enterococcal infections. Expert Rev Anti
guideline for the use of antimicrobial agents in neutropenic dime as empiric therapy for fever in neutropenic patients Infect Ther. 2008;6:637-655.
patients with cancer: 2010 update by the Infectious Dis- with cancer. Ann Pharmacother. 2000;34:989-995. 298. Lagace-Wiens PR, Tailor F, Simner P, et al. Activity of
eases Society of America. Clin Infect Dis. 2011;52:e56-e93. 275. Cordonnier C, Herbrecht R, Pico JL, et al. Cefepime/ NXL104 in combination with beta-lactams against geneti-
250. Nicolau D, McNabb J, Lacey M. Continuous versus inter- amikacin versus ceftazidime/amikacin as empirical therapy cally characterized Escherichia coli and Klebsiella pneu-
mittent administration of ceftazidime in intensive care unit for febrile episodes in neutropenic patients: a comparative moniae isolates producing class A extended-spectrum
patients with nosocomial pneumonia. Int J Antimicrob study. Clin Infect Dis. 1997;24:41-51. beta-lactamases and class C beta-lactamases. Antimicrob
Agents. 2001;17:497-504. 276. Kieft H, Hoepelman AIM, Arska MR, et al. Cefepime com- Agents Chemother. 2011;55:2434-2437.
251. Lorente L, Jimenez A, Palermo S, et al. Comparison of pared with ceftazidime as initial therapy for serious bacte- 299. Walkty A, DeCorby M, Lagace-Wiens PR, et al. In vitro
clinical cure rates in adults with ventilator-associated rial infections and sepsis syndrome. Antimicrob Agents activity of ceftazidime combined with NXL104 versus
pneumonia treated with intravenous ceftazidime adminis- Chemother. 1994;38:415-421. Pseudomonas aeruginosa isolates obtained from patients in
tered by continuous or intermittent infusion: a retrospec- 277. Sanders WE Jr, Tenney JH, Kessler RE. Efficacy of cefepime Canadian hospitals (CANWARD 2009 study). Antimicrob
tive, nonrandomized, open-label, historical chart review. in the treatment of infections due to multiply resistant Agents Chemother. 2011;55:2992-2994.
Clin Ther. 2007;29:2433-2439. Enterobacter species. Clin Infect Dis. 1996;23:454-461. 300. Mushtaq S, Warner M, Livermore DM. In vitro activity of
252. Blumer JL, Stern RC, Klinger JD, et al. Ceftazidime therapy 278. Barckow D, Schwigon CD. Cefepime versus cefotaxime in ceftazidime+NXL104 against Pseudomonas aeruginosa and
in patients with cystic fibrosis and multiply-drug-resistant the treatment of lower respiratory tract infections. J Anti- other non-fermenters. J Antimicrob Chemother. 2010;65:
pseudomonas. Am J Med. 1985;79(suppl 2A):37-46. microb Chemother. 1993;32(suppl B):187-193. 2376-2381.
253. Fong IW, Tompkins KB. Review of Pseudomonas aerugi- 279. Zervos M, Nelson M, Cefepime Study Group. Cefepime 301. Zhanel GG, Lawson CD, Adam H, et al. Ceftazidime-
nosa meningitis with special emphasis on treatment with versus ceftriaxone for empiric treatment of hospitalized avibactam: a novel cephalosporin/β-lactamase inhibitor
ceftazidime. Rev Infect Dis. 1985;7:604-612. patients with community-acquired pneumonia. Antimicrob combination. Drugs. 2013;73159-73177.
254. Handsfield HH, McCormack WM, Hook EW, et al. A com- Agents Chemother. 1998;42:729-733. 302. Mushtaq S, Warner M, Williams G, et al. Activity of che-
parison of single-dose cefixime with ceftriaxone as treat- 280. Saez-Llorens X, Castano E, Garcia R, et al. Prospective querboard combinations of ceftaroline and NXL104 versus
ment for uncomplicated gonorrhea. N Engl J Med. 1991; randomized comparison of cefepime and cefotaxime for beta-lactamase-producing Enterobacteriaceae. J Antimi-
325:1337-1341. treatment of bacterial meningitis in infants and children. crob Chemother. 2010;65:1428-1432.
255. Asbach HW. Single dose oral administration of cefixime Antimicrob Agents Chemother. 1995;39:937-940. 303. Sader HS, Flamm RK, Jones RN. Antimicrobial activity of
400 mg in the treatment of acute uncomplicated cystitis 281. Saez-Llorens X, O’Ryan M. Cefepime in the empiric treat- ceftaroline-avibactam tested against clinical isolates col-
and gonorrhoea. Drugs. 1991;42(suppl 4):10-13. ment of meningitis in children. Pediatr Infect Dis J. 2001; lected from U.S. medical centers. Antimicrob Agents Che-
256. Centers for Disease Control and Prevention. Update to 20:356-361. mother. 2013;57:1982-1988.
CDC’s sexually transmitted diseases treatment guidelines, 282. Paul M, Yahav D, Fraser A, Leibovici L. Empirical antibiotic 304. Vazquez JA, Gonzalez Patzan LD, Stricklin D, et al. Efficacy
2010: oral cephalosporins no longer a recommended treat- monotherapy for febrile neutropenia: systematic review and safety of ceftazidime-avibactam versus imipenem-
ment for gonococcal infections. MMWR Morb Mortal and meta-analysis of randomized controlled trials. J Anti- cilastatin in the treatment of complicated urinary tract
Wkly Rep. 2012;61:590-594. microb Chemother. 2006;57:176-189. infections, including acute pyelonephritis, in hospitalized
257. Wormser GP, Dattwyler RJ, Shapiro ED, et al. The clinical 283. Yahav D, Paul M, Fraser A, et al. Efficacy and safety of adults: results of a prospective, investigator-blinded, ran-
assessment, treatment, and prevention of Lyme disease, cefepime: a systematic review and meta-analysis. Lancet domized study. Curr Med Res Opin. 2012;28:1921-1931.
human granulocytic anaplasmosis, and babesiosis: clinical Infect Dis. 2007;7:338-348. 305. Lucasti B, Popescu I, Ramesh MK, et al. Comparative study
practice guidelines by the Infectious Diseases Society of 284. Towne TG, Lewis JS, Echevarria K. Efficacy and safety of of the efficacy and safety of ceftazidime/avibactam plus
America. Clin Infect Dis. 2006;43:1089-1134. cefepime. Lancet Infect Dis. 2009;9:4-5. metronidazole versus meropenem in the treatment of com-
258. Augenbraun M, Workowski K. Ceftriaxone therapy for 285. Kim PW, Wu YT, Cooper B, et al. Meta-analysis of a pos- plicated intra-abdominal infections in hospitalized adults:
syphilis: report from the emerging infections network. Clin sible signal of increased mortality associated with cefepime results of a randomized, double-blind, phase II trial. J Anti-
Infect Dis. 1999;29:1337-1338. use. Clin Infect Dis. 2010;51:381-389. microb Chemother. 2013;68:1183-1192.
259. Guerrant RL, van Gilder T, Steiner TS, et al. Practice guide- 286. McGee L, Biek D, Ge Y, et al. In vitro evaluation of the 306. Bulik CC, Christensen H, Nicolau DP. In vitro potency of
lines for the management of infectious diarrhea. Clin Infect antimicrobial activity of ceftaroline against cephalosporin- CXA-101, a novel cephalosporin, against Pseudomonas
Dis. 2001;32:331-351. resistant isolates of Streptococcus pneumoniae. Antimicrob aeruginosa displaying various resistance phenotypes,
260. Finch RG. Third-generation cephalosporins in the treat- Agents Chemother. 2009;53:522-526. including multidrug resistance. Antimicrob Agents Che-
ment of rare infections. Am J Med. 1990;88(Suppl 4A): 287. Kosowska K, Hoellman DB, Gengrong L, et al. Antipneu- mother. 2010;54:557-559.
25S-31S. mococcal activity of ceftobiprole, a novel broad-spectrum 307. Sader HS, Rhomberg PR, Farrell DJ, Jones RN. Antimicro-
261. Wilson WR, Karchmer AW, Dajani AS, et al. Antibiotic cephalosporin. Antimicrob Agents Chemother. 2005;49: bial activity of CXA-101, a novel cephalosporin testing
treatment of adults with infective endocarditis due to strep- 1932-1942. combination with tazobactam against Enterobacteriaceae,
tococci, enterococci, staphylococci, and HACEK microor- 288. Noel GJ, Bush K, Bagchi P, et al. A randomized, double- Pseudomonas aeruginosa, and Bacteroides fragilis strains
ganisms. JAMA. 1995;274:1706-1713. blind trial comparing ceftobiprole medocaril with vanco- having various resistance phenotypes. Antimicrob Agents
262. Simmons G, Jones N, Calder L. Equivalence of ceftriaxone mycin plus ceftazidime for the treatment of patients with Chemother. 2011;55:2390-2394.
and rifampicin in eliminating nasopharyngeal carriage of complicated skin and skin-structure infections. Clin Infect 308. Moya B, Zamorano L, Juan C, et al. Affinity of the new
serogroup B Neisseria meningitidis. J Antimicrob Che- Dis. 2008;46:647-655. cephalosporin CXA-101 to penicillin-binding proteins of
mother. 2000;45:909-911. 289. Deresinski SC. The efficacy and safety of ceftibiprole in the Pseudomonas aeruginosa. Antimicrob Agents Chemother.
263. Martin MJ, Hutchinson NA, Eltringham IJ, et al. Secondary treatment of complicated skin and skin structure infec- 2010;54:3933-3937.
prevention of meningococcal disease: ceftriaxone or cipro- tions: evidence from 2 clinical trials. Diagn Microbiol Infect 309. Craig WA, Andes DR. In vivo activities of ceftolozane, a
floxacin should be considered as first line prophylaxis. Dis. 2008;61:103-109. new cephalosporin, with and without tazobactam against
BMJ. 1996;312:1536-1537. 290. Corey GR, Wilcox M, Talbot GH, et al. Integrated analysis Pseudomonas aeruginosa and Enterobacteriaceae, includ-
264. Bradsher RW, Snow RM. Ceftriaxone treatment of skin and of CANVAS 1 and 2: phase 3, multicenter, randomized, ing strains with extended-spectrum β-lactamases, in the
soft tissue infections in a once daily regimen. Am J Med. double-blind studies to evaluate the safety and efficacy of thighs of neutropenic mice. Antimicrob Agents Chemother.
1984;77(suppl C):63-67. ceftaroline versus vancomycin plus aztreonam in compli- 2013;57:1577-1582.
265. Duncan CJ, Barr DA, Seaton RA. Outpatient parenteral cated skin and skin-structure infections. Clin Infect Dis. 310. Livermore DM, Mushtaq S, Ge Y. Chequerboard titration
antimicrobial therapy with ceftriaxone, a review. Int J Clin 2010;51:641-650. of cephalosporin CXA-101 (FR264205) and tazobactam
Pharm. 2012;34:410-417. 291. Nicholson SC, Welte T, File TM Jr, et al. A randomized, versus beta-lactamase-producing Enterobacteriaceae.
266. Sexton DJ, Tenenbaum MJ, Wilson WR, et al, Endocarditis double-blind trial comparing ceftobiprole medocaril with J Antimicrob Chemother. 2010;65:1972-1974.
Treatment Consortium Group. Ceftriaxone once daily for ceftriaxone with or without linezolid for the treatment of

You might also like