DETERMINATION OF ISOLATE

PATHOGENICITY

CASE 62-1, QUESTION 7: Serratia marcescens grows from a culture of R.G.’s endotracheal aspirate. How

can it be determined whether an isolate represents a true bacterial infection versus colonization or

contamination?

A positive culture may represent colonization, contamination, or infection.

Colonization indicates that bacteria are present at the site; however, they are not

actively causing infection. Poor sampling techniques or inappropriate handling of

specimens can result in contamination. Contamination differs from colonization in

that these isolates are not truly at the site in question. The S. marcescens growing

from R.G. could represent infection, colonization, or contamination. If a suction

catheter was used to sample R.G.’s endotracheal aspirate, the infecting organism

likely would be cultured; however, other nonpathogenic flora would also appear in

the culture medium (colonization). Furthermore, if the sample is not handled

aseptically by the clinician or the microbiology laboratory, bacterial contamination is

possible.

In summary, culture results do not solely identify true pathogens. In R.G., the

Serratia may be a pathogen, contaminant, or colonizer. Nevertheless, considering the

severity of R.G.’s illness and his associated respiratory symptoms, treatment directed

against this pathogen is necessary.

ANTIMICROBIAL TOXICITIES

CASE 62-1, QUESTION 8: In light of the positive culture for Serratia, his increased respiratory secretions,

and a worsening chest radiograph, ventilator-associated pneumonia is likely present. Pending susceptibility

results, R.G. is empirically started on imipenem and gentamicin. In review of his patient records, R.G. has no

known allergies. Are there equally effective, less toxic options for this patient?

p. 1331

p. 1332

Adverse Effects and Toxicities

Before antimicrobial therapy is started, it is important to elicit an accurate drug and

allergy history. When “allergy” has been reported by the patient, it is necessary to

determine whether the reaction was intolerance, toxicity, or true allergy. For

example, gastric intolerance caused by oral doxycycline is common; however, this

adverse effect does not represent an allergic manifestation. Although R.G. has no

known allergies, neither imipenem nor gentamicin are optimal choices. Imipenem is

associated with seizures, particularly in patients with renal failure and in doses in

excess of 50 mg/kg/day. Considering R.G.’s acute onset of renal failure and his

history of seizures, other carbapenems, such as meropenem or doripenem, or

alternative classes of antibacterials would be preferable. Gentamicin similarly may

not be a good choice in R.G. His increased age and declining renal function

predispose him to aminoglycoside nephrotoxicity and ototoxicity (cochlear and

vestibular).

21 A reasonable recommendation pending susceptibility results would be

to discontinue imipenem and gentamicin and treat with meropenem or doripenem

with or without a fluoroquinolone. Table 62-8 lists common antibiotics adverse

effects and toxicities.

p. 1332

p. 1333

Table 62-8

Antibiotic Adverse Effects and Toxicities

Antibiotics Side Effects Comments

β-Lactams

(penicillin,

cephalosporins,

monobactams,

penems)

Allergic: anaphylaxis,

urticaria, serum

sickness, rash, fever

Many patients will have “ampicillin rash” or “β-lactam rash” with

no cross-reactivity with any other penicillins/β-lactams. Most

commonly observed in patients with concomitant EBV disease.

Likelihood of IgE-mediated cross-reactivity between penicillins and

cephalosporins approximately 5%–10%. Most recent data strongly

suggest minimal IgE cross-reactivity between penicillins and

imipenem/meropenem. No IgE cross-reactivity between

aztreonam and penicillins.

Diarrhea Particularly common with ampicillin, augmentin, ceftriaxone.

Antibiotic-associated colitis can occur with most antimicrobials.

Hematologic:

anemia,

thrombocytopenia,

Hemolytic anemia more common with higher doses. Antiplatelet

activity (inhibition of platelet aggregation) most common with the

antipseudomonal penicillins and high serum levels of other β-

antiplatelet activity,

hypothrombinemia

lactams.

Hypothrombinemia more often associated with those

cephalosporins with the methyltetrazolethiolside chain

(cefamandole, cefotetan). Reaction preventable and reversible

with vitamin K.

Hepatitis or biliary

sludging

Hepatitis most common with oxacillin. Biliary sludging and stones

reported with ceftriaxone.

Phlebitis

Seizure activity Associated with high levels of β-lactams, particularly penicillins

and imipenem.

Potassium load Penicillin G (K+).

Nephritis Occasionally reported for most β-lactams.

Neutropenia Nafcillin.

Disulfiram reaction Associated with cephalosporins with methyltetrazolethiolside chain

(cefamandole, cefotetan).

Hypotension, nausea Associated with fast infusion of imipenem.

Aminoglycosides

(gentamicin,

tobramycin,

amikacin, netilmicin)

Nephrotoxicity Average 10%–15% incidence. Generally reversible, usually occurs

after 5–7 days of therapy. Risk factors: dehydration, age, dose,

duration, concurrent nephrotoxins, liver disease.

Ototoxicity 1%–5% incidence, often irreversible. Both cochlear and vestibular

toxicity occur.

Neuromuscular

paralysis

Rare, most common with large doses administered via

intraperitoneal instillation or in patients with myasthenia gravis.

Macrolides

(erythromycin,

azithromycin,

clarithromycin)

Nausea, vomiting,

“burning” stomach,

cholestatic jaundice,

ototoxicity, prolonged

QT interval

Oral administration. Azithromycin and clarithromycin associated

with less nausea than erythromycin.

Cholestatic jaundice reported for all erythromycin salts, most

common with estolate.

Ototoxicity most common with high doses in patients with renal or

hepatic failure.

Torsades de Pointes and increased risk of cardiac death

associated with increased QT interval.

p. 1333

p. 1334

Telithromycin Hepatotoxicity;

upper GI

Severe, sometime fatal hepatotoxicity associated with

telithromycin.

Clindamycin Diarrhea Most common adverse effect. High association with antibioticassociated colitis.

Tetracyclines

(including tigecycline)

Allergic

Photosensitivity

Drug Interactions ↓ Oral bioavailability with multivalent cations.” similar to that

stated in the Quinolones section

Teeth and bone

deposition and

discoloration

Avoid in pediatrics (<8 years old), pregnancy, and breastfeeding.

GI Upper GI predominates.

Hepatitis Primarily in pregnancy or the elderly. Renal (azotemia) Tetracyclines have antianabolic effect and should be avoided in

patients with ↓ renal function. Less problematic with

doxycycline.

Vestibular Associated with minocycline, particularly high doses.

Vancomycin “Red Man

Syndrome”:

hypotension, flushing

Associated with rapid infusion of vancomycin. More common

with increased doses.

Nephrotoxicity Reversible nephrotoxicity with high doses or in combination with

other nephrotoxins.

Ototoxicity Only with receipt of concomitant ototoxins such as

aminoglycosides or macrolides.

Phlebitis Needs large volume dilution.

Dalfopristin/quinupristin Phlebitis Generally requires central line administration.

Myalgia Moderate to severe in many patients.

Increased bilirubin

Daptomycin Myalgia Primarily at high doses and reversible.

Linezolid (Tedizolid) Thrombocytopenia,

neutropenia, anemia,

MAO inhibition,

neuropathy

Bone marrow suppression and neuropathy duration- and dosedependent with linezolid. Tedizolid may have less association

with bone marrow suppression and neuropathy.

Dalbavancin,

Oritavancin,

Telavancin

Renal toxicity and

prolonged QT with

telavancin

Sulfonamides GI Nausea, diarrhea.

Hepatic Cholestatic hepatitis, ↑ incidence in HIV.

Rash

Hyperkalemia

Exfoliative dermatitis, Stevens–Johnson syndrome. More

common in HIV.

Only with trimethoprim (as a component of trimethoprim–

sulfamethoxazole).

Bone marrow Neutropenia, thrombocytopenia. More common in HIV.

Kernicterus Caused by unbound drug in the neonate. Premature liver cannot

conjugate bilirubin. Sulfonamide displaces bilirubin from protein,

resulting in excessive free bilirubin and kernicterus.

Chloramphenicol Anemia Idiosyncratic irreversible aplastic anemia (rare). Reversible

dose-related anemia.

Gray syndrome Caused by inability of neonates to conjugate chloramphenicol.

Quinolones GI Nausea, vomiting, diarrhea.

Prolonged QT Moxifloxacin; possibly all quinolones as a class.

Drug interactions ↓ Oral bioavailability with multivalent cations.

CNS Altered mentalstatus, confusion, seizures.

Cartilage toxicity Toxic in animal model. Despite this toxicity, appears safe in

children including patients with cystic fibrosis.

Tendonitis or tendon

rupture

Common in elderly, renal failure, concomitant glucocorticoids.

Antifungals

Amphotericin B

products

Nephrotoxicity Common. May depend on patient sodium load. Caution with

concomitant nephrotoxins (e.g., aminoglycosides, cyclosporine).

Hypokalemia Predictable. Probably caused by renal tubular excretion of

potassium. More common in patients receiving concomitant

piperacillin–tazobactam.

Hypomagnesemia Less commonly observed than hypokalemia.

Anemia Long-term adverse effect. Similar to anemia of chronic disease.

Caspofungin,

Micafungin,

Anidulofungin

Mild LFT increase

with concomitant

cyclosporine

Anidulofungin is reconstituted with alcohol (about the equivalent of

a beer).

Flucytosine Neutropenia,

thrombocytopenia

Secondary to metabolism of flucytosine to fluorouracil. More

commonly observed with flucytosine levels >100 mg/mL. More

common in patients with HIV.

Hepatitis Usually moderate ↑ in LFT. Rarely clinical hepatitis.

Ketoconazole

(fluconazole,

isavuconazole,

itraconazole,

posaconazole,

voriconazole)

Drug interactions ↓ Oral bioavailability of ketoconazole tablet, and itraconazole

capsules with ↑ gastric pH. Azoles are CYP450 substrates and

also inhibitors of CYP450 3A4 and other CYP isoenzymes.

Voriconazole most likely to be associated with CYP drug

interactions

Hepatitis Ranges from mild ↑ in LFT to occasional fatal hepatitis.

Gynecomastia, ↓

libido

More common with high-dose ketoconazole (>400 mg/day). Less

common with other azoles.

Visual disturbance Unique to voriconazole, particularly first week of therapy.

Antivirals (Excluding Antiretrovirals and Hepatitis Antivirals)

Acyclovir Phlebitis Caused by poor solubility of IV preparation. Reported in 1%–20%

of cases.

Renal failure Low solubility of acyclovir associated with renal failure.

Dehydrated patients, as well as rapid infusions, predispose to

toxicity.

CNS 1% incidence in AIDS. ↑ Incidence with dose in >10 mg/kg/day.

Foscarnet Nephrotoxicity Occurs in up to 60% of patients. May be prevented with normal

saline bolus before dose. Frequent monitoring of renal function

imperative.

Mineral and

electrolyte

abnormalities

↑ and ↓ calcium or phosphate may be observed. Hypocalcemia,

hypo- and hyperphosphatemia, hypomagnesemia, hypokalemia. ↑

Risk of cardiomyopathy and seizures.

Anemia Anemia in 33%; usually manageable with transfusions and

discontinuation of foscarnet.

Nausea, vomiting

Ganciclovir Neutropenia,

thrombocytopenia

↑ Incidence in AIDS. ↑ Incidence with doses in excess of 10

mg/kg/day.

Hepatitis Usually mild to moderate in LFT.

Oseltamavir Nausea

AIDS, acquired immunodeficiency syndrome; CNS, central nervous system; EBV, Epstein–Barr virus; GI,

gastrointestinal; HIV, human immunodeficiency virus; IV, intravenous; LFT, liver function tests; MAO,

monoamine oxidase.

Concomitant Disease States

Concomitant disease states should also be considered in the selection of therapy. As

discussed above, older patients with hearing deficits are poor candidates for

potentially ototoxic aminoglycoside therapy. Diabetic or kidney transplant patients

with candidemia may be better treated with fluconazole or an echinocandin rather

than nephrotoxic amphotericin B products. Patients with a preexisting seizure history

should not receive imipenem if less toxic therapy can be used. In summary, the

toxicologic profile of a drug must be taken into account in the selection of

antimicrobial therapy.

p. 1334

p. 1335

ANTIMICROBIAL COSTS OF THERAPY

CASE 62-1, QUESTION 9: What factors should be included in calculating the cost of R.G.’s antimicrobial

therapy?

The true cost of antimicrobial therapy is difficult to quantify.

22 Although

acquisition cost traditionally has been the primary factor in the overall cost of

therapy, drug administration labor costs (i.e., nursing and pharmacy) and the use of

IV sets, piggyback bags, and infusion control devices must be included in the

analysis. As a result, a drug that must be administered several times daily, such as

intravenous penicillin, will incur increased administration costs compared with one,

such as ceftriaxone, that requires once-a-day dosing.

Some drugs, such as aminoglycosides, are associated with increased laboratory

costs (e.g., aminoglycoside serum concentrations, serum creatinine, and audiometry)

that are not required for other agents,

23 such as the third-generation cephalosporins

and quinolones. Similarly, drugs with a high potential for misuse or toxicity can be

associated with increased costs because of monitoring (e.g., medication use

evaluation, pharmacokinetic monitoring). If meropenem with or without ciprofloxacin

had been selected for R.G., this therapy would be expected to be associated with

relatively few laboratory costs. However, the broad spectrum of activity

24 of these

agents, potential for misuse, and development of resistance might, however, result in

increased monitoring costs and overall cost to society.

Costs that are difficult to quantitate include those associated with failure of

antimicrobial therapy and antimicrobial toxicity. Ineffective or toxic therapy can

prolong hospitalization and may require expensive interventions, such as

hemodialysis,

23 mechanical ventilation, and intensive care unit admission. The net

effect of these latter costs can be significantly greater than the acquisition and

administration costs of antimicrobial therapy.

In summary, determining the true cost of antimicrobial therapy is complex.

Acquisition cost, IV bags, infusion controllers, and labor must be incorporated into

the analysis. Although they are difficult to estimate, other costs, including antibiotic

toxicity and failure of therapy, should also be included.

ROUTE OF ADMINISTRATION

CASE 62-1, QUESTION 10: The Serratia was determined to be susceptible to ciprofloxacin. Oral

ciprofloxacin was considered for the treatment of R.G.’s presumed Serratia pneumonia, but the IV route was

prescribed. Why is the oral administration of ciprofloxacin reasonable (or unreasonable) in R.G.?

The proper route of antibiotic administration depends on many factors, including

the severity of infection, antimicrobial oral bioavailability, and other patient factors.

In patients who appear “septic,” blood flow often is shunted away from the

mesentery and extremities, resulting in unreliable bioavailability from the

gastrointestinal (GI) tract or muscles. Consequently, hemodynamically unstable

patients should always receive antimicrobials by the IV route to ensure therapeutic

antimicrobial levels. Furthermore, some drug interactions with oral agents can result

in subtherapeutic serum concentrations (e.g., reduced bioavailability associated with

concomitant quinolone and antacid administration and the decreased absorption of

itraconazole with concurrent proton-pump inhibitor [PPI] therapy).

R.G. is clinically septic with a possible Serratia pneumonia. Considering his

unstable state, the bioavailability of oral ciprofloxacin cannot be guaranteed; thus, he

should be treated with IV antimicrobials.

ANTIMICROBIAL DOSING

CASE 62-1, QUESTION 11: What dose of IV ciprofloxacin should be given to R.G.? What factors must be

taken into account in determining a proper antimicrobial dose?

The choice of dosing regimen is based on many factors. Table 62-9 provides a

guide for the dosing of more commonly administered antimicrobials. Selection of the

appropriate dosage should be based on evidence confirming the efficacy of the

dosage in the treatment of a specific infection. Patient-specific factors, including

weight, site of infection, and route of elimination, must also be considered in dosage

selection. The patient’s weight is important, particularly for agents with a low

therapeutic index (e.g., aminoglycosides, imipenem, and flucytosine); these drugs

should be dosed on a milligram per kilogram per day basis. Other agents with a more

favorable adverse effect profile, such as cephalosporins, are less likely to require

weight-specific dosing in most disease states.

Site of Infection

Site of infection results in different dosage requirements. An uncomplicated urinary

tract infection requires lower doses considering the high urinary drug concentrations

that are achieved with most renally cleared agents. In contrast, a more serious upper

urinary tract infection, such as pyelonephritis, requires increased doses to ensure

therapeutic drug levels in tissue and in serum.

Anatomic and Physiologic Barriers

Anatomic and physiologic barriers must also be considered in evaluating a dosing

regimen. For example, penetration into cerebrospinal fluid requires high doses to

ensure adequate antimicrobial concentrations.

25 Vitreous humor

26 and the prostate

gland

27 are additional sites in which therapeutic antimicrobial concentrations are

more difficult to achieve.

Route of Elimination

Route of elimination must also be considered in the dosage calculation. In general,

antimicrobials are eliminated renally or nonrenally (metabolic or biliary). Renal

function can be estimated via 24-hour urine collection or with equations, such as the

Cockcroft and Gault equation

28

:

Several anti-infectives are eliminated renally (Table 62-9). Most β-lactams are

eliminated by the kidney. In contrast, ceftriaxone and most antistaphylococcal

penicillins (e.g., nafcillin, oxacillin, and dicloxacillin) are eliminated both renally

and nonrenally. Aminoglycosides, vancomycin, acyclovir, and ganciclovir are

cleared primarily by the kidney. Thus, dosage adjustment is recommended for these

drugs in patients with renal failure (Table 62-9). Because azithromycin, clindamycin,

and metronidazole are primarily eliminated by the liver, no dose reduction is

required in renal failure for these agents. Using the Cockcroft and Gault equation,

R.G.’s age (63 years), weight (70 kg), and current serum creatinine (3.8 mg/dL)

results in a calculated creatinine clearance of 14 mL/minute. R.G. normally would be

given an IV dosage of ciprofloxacin at 400 mg every 12 hours. His increasing

creatinine, however, suggests that his dosage should be decreased to 200 to 300 mg

every 12 hours.

p. 1335

p. 1336

Table 62-9

Adult Antimicrobial Dosing Guidelines for Hospitalized Patients (Selected

Drugs)

Drug

CrCl > 50

mL/minute CrCl 10–50 mL/minute

CrCl <10

mL/minute

(ESRD not

on HD)

Dialysis

(HD or

CRRT)

Acyclovir Herpes simplex

infections

5 mg/kg/dose IV every

8 hours

5 mg/kg/dose IV every 12–24

hours

2.5 mg/kg IV

every 24

hours

HD: 2.5

mg/kg IV ×1

now then 2.5

mg/kg every

evening (give

after HD on

HD days)

CRRT: 5

mg/kg every

24 hours

HSV

encephalitis/Herpes

zoster

10 mg/kg/dose IV

every 8 hours

10 mg/kg/dose IV every 12–

24 hours

5 mg/kg IV

every 24

hours

HD: 5 mg/kg

IV ×1 now

then 5 mg/kg

every evening

(give after

HD on HD

days)

CRRT: 5–10

mg/kg every

12–24 hours

Ampicillin Meningitis or

endovascular infection

2 g IV every 4 hours

Uncomplicated

infection

2 g IV every 6 hours

2 g IV every 6 hours 1 g IV every

8–12 hours

HD: 1–2 g IV

every 12

hours

CRRT: 1–2 g

IV every 6

hours

Ampicillin/sulbactam 3 g IV every 6 hours 1.5 g IV every 6 hours 1.5 g IV

every 12

hours

HD: 1.5–3 g

IV every 12

hours

CRRT: 1.5 g

IV every 6

hours

Aztreonam 2 g IV every 8 hours 2 g IV every 12 hours 1 g IV every

12 hours

HD: 1 g IV

×1 now then

1 g every

evening (give

after HD on

HD days)

CRRT: 2 g

IV every 12

hours

Cefazolin Gram negative or

complicated Gram

1–2 g IV every 12 hours 1 g IV every

24 hours

HD: 2 g after

HD only

positive

2 g IV every 8 hours

Uncomplicated Gram

positive

1–2 g IV every 8 hours

CRRT: 2 g

IV every 12

hours

Caspofungin

Severe hepatic

dysfunction: 70 mg

LD, then 35 mg IV

daily

LD: 70 mg × 1, then 50

mg every 24 hours

Increase maintenance

dose to 70 mg when

given with phenytoin,

rifampin,

carbamazapine,

dexamethasone,

nevirapine, efavirenz

No change No change No change

Cefepime

Less severe infections

Febrile neutropenia,

meningitis,

pseudomonas

infections, critically ill

patients

>60 mL/minute

2 g IV every 12 hours

2 g IV every 8 hours

30–60

mL/minute

2 g IV every

24 hours

2 g IV every

12 hours

10–30

mL/minute

1 g IV every

24 hours

2 g IV every

24 hours

<10

mL/minute

500 mg IV

every 24

hours

1 g IV every

24 hours

HD: 2 g after

HD only

CRRT: 2 g

IV every 12

hours

Ceftazidime 2 g IV every 8 hours 2 g IV every 12–24 hours 500 mg IV

every 24

hours

HD: 1 g after

HD only

CRRT: 2 g

IV every 12

hours

Ceftriaxone

Meningitis: 2 g every

12 hours

Endocarditis and

osteomyelitis: 2 g

every 24 hours

1 g IV every 24 hours No change No change No change

p. 1336

p. 1337

Ciprofloxacin

IVPO

Pseudomonas

infections

400 mg IV every

12 hours

500–750 mg PO every 12

hours

400 mg IV every 8 hours

750 mg PO every 12 hours

30–50

mL/minute

No change

No change

10–30

mL/minute

200–400 mg

IV every 12

hours

250–500 mg

PO every 12

hours

< 10

mL/minute

200 IV every

12 hours

250 mg PO

every 12

hours

HD: 400 mg

IV every 24

hours or 500

mg PO every

24 hours (give

after HD on

HD days)

CRRT: 400

mg IV every

12 hours

Clindamycin 600–900 mg IV every 8

hours

No change No change No change

Daptomycin

Not effective in

the treatment of

4–10 mg/kg IV every 24

hours

Dose depends on indication

<30 mL/minute

4–10 mg/kg IV every 48 hours

HD: 4–10

mg/kg IV

every 48

pneumonia hours

CRRT: 4–10

mg/kg IV

every 48

hours

Doxycycline

IVPO

100 mg IV/PO every 12

hours

No change No change No change

Ertapenem 1 g IV every 24 hours <30 mL/minute

500 mg IV every 24 hours

HD: 500 mg

every 24

hours

CRRT: 500

mg every 24

hours

Ethambutol 15–20 mg/kg PO every 24

hours

<30 mL/minute

15–25 mg/kg 3 times/week

HD: 15–25

mg/kg 3

times/week

(after HD)

CRRT: 15–25

mg/kg 3

times/week

Fluconazole

IVPO

Oropharyngeal candidiasis

100 mg every 24 hours

Esophageal candidiasis

200 mg every 24 hours

Severe infection

Loading dose of 800 mg then

400 mg every 24 hours

50–200 mg IV/PO every 24

hours

50–100 mg

IV/PO every

24 hours

HD: 400 mg

after HD only

CRRT: 400–

800 mg every

24 hours

Flucytosine

(5FC)

Meningitis

25 mg/kg/dose PO every 6

hours

25–50

mL/minute

25

mg/kg/dose

PO every 12

hours

10–25

mL/minute

25

mg/kg/dose

PO every 24

hours

12.5

mg/kg/dose

PO every 24

hours

HD: 12.5–25

mg/kg PO

every 24

hours

CRRT: 12.5–

37.5

mg/kg/dose

PO every 12–

24 hours

Ganciclovir >70 mL/minute

5 mg/kg/dose IV every 12

hours

50–69

mL/minute

2.5

mg/kg/dose

IV every 12

hours

25–49

mL/minute

2.5 mg/kg IV

every 24

hours

10–24

mL/minute

1.25 mg/kg

IV every 24

hours

HD: 1.25

mg/kg after

HD only

CRRT: 2.5

mg/kg every

24 hours

Gentamicin See Tobramycin See

Tobramycin

See Tobramycin See

Tobramycin

Imipenem 500 mg IV every 6–8 hours

max 50 mg/kg/day

500 mg IV every 8 hours <20

mL/minute

250–500 mg

IV every 12

hours

HD: 250 mg

IV every 12

hours

CRRT: 500

mg IV every

8 hours

Isoniazid 300 mg PO every 24 hours No change No change No change

p. 1337

p. 1338

Levofloxacin

IV-PO

Urinary tract infection

Pneumonia

Pseudomonas

infections

250–500 mg IV/PO

every 24 hours

750 mg IV/PO every

24 hours

500 mg ×1, then 250 mg

IV/PO every 24 hours

750 mg ×1; then 750 IV/PO

every 48 hours

500 mg ×1,

then 250 mg

IV/PO every

48 hours

750 mg ×1,

then 500 mg

IV/PO every

48 hours

HD: 500 mg

×1, then 250

mg every 48

hours

CRRT: 500

mg ×1, then

250–500 mg

every 24

hours

Linezolid

IV-PO 600 mg IV/PO every

12 hours

No change No change No change

Meropenem

Meningitis,

documented or

suspected

Pseudomonas

infections or critically

ill

0.5–1 g IV every 8

hours

2 g IV every 8 hours

25–50

mL/minute

0.5–1 g IV

every 12

hours

2 g IV every

12 hours

10–25

mL/minute

0.5 g IV

every 12

hours

1 g IV every

12 hours

0.5 g IV

every 24

hours

1 g IV every

24 hours

HD: 500 mg

IV ×1 now

then 500 mg

every evening

(give after

HD on HD

days)

CRRT: 1 g

IV every 12

hours

Metronidazole

IV-PO 500 mg IV/PO every 8

hours

500 mg IV/PO every 8 hours 500 mg

IV/PO every

12 hours

ESRD not on

HD

500 mg

IV/PO every

8 hours

Moxifloxacin

IV-PO 400 mg IV/PO every

24 hours

No change No change No change

Nafcillin

Meningitis,

osteomyelitis,

endovascular infection

1–2 g IV every 4–6

hours

2 g IV every 4 hours

No change No change No change

Penicillin G

Meningitis,

endovascular infection

2–3 MU IV every 4–6

hours

3–4 MU IV every 4–6

hours

1–2 MU IV every 4–6 hours 1 MU IV

every 6 hours

HD: 1 MU

IV every 6

hours

CRRT: 2 MU

IV every 4–6

hours

Piperacillin/tazobactam

(Zosyn)

Documented/suspected

Pseudomonas

infections

3.375–4.5 g IV every

6–8 hours

4.5 g every 6 hours for

CrCl >20 mL/minute

3.375–4.5 g every 6–8 hours 2.25 g every

8 hours

HD: 2.25 g

IV every 8

hours

CRRT: 4.5 g

IV every 8

hours or

3.375 g IV

every 6 hours

Posaconazole

Must be administered

with high-fat meal or

400 mg PO every 12

hours or

200 mg PO every 6

No change No change No change

nutritionalshake, e.g.,

Ensure

Neutropenia

prophylaxis

hours

200 mg PO every 8

hours

Pyrazinamide 20–25 mg/kg/day PO

every 24 hours

<30 mL/minute

25–35 mg/kg 3 times/week

HD: 25–35

mg/kg 3

times/week

after HD

CRRT: 25–35

mg/kg 3

times/week

Rifampin

Mycobacterial

infection

Endocarditis

Prosthetic device

infection

600 mg PO every 24

hours

300 mg PO every 8

hours

450 mg PO every 12

hours

No change No change No change

Tigecycline

Severe hepatic disease:

100 mg IV ×1, then 25

mg IV every 12 hours

100 mg IV × 1, then 50

mg IV every 12 hours

No change No change No change

p. 1338

p. 1339

Tobramycin

(and

gentamicin)

Gram negative

infection

Dose is based on

ideal body

weight (IBW)

except in obese

patients or those

under their IBW.

Use actual body

weight if patient

weight is less

than IBW. Use

adjusted body

weight if patient

is obese.

(calculations see

below)

For CrCl > 60 mL/minute, not

morbidly obese or fluid

overloaded, give once daily

regimen:

7 mg/kg/dose IV every 24

hours.

For those patients not

qualifying for once daily

dosing, see conventional

dosing below.

For conventional

dosing peak and

trough levels

should be

monitored.

>60 mL/minute

1.6 mg/kg IV every 8 hours

40–60 mL/minute

1.2–1.5 mg/kg every 12–24

hours

<20

mL/minute

2 mg/kg

loading dose,

then follow

drug levels

HD: 2 mg/kg

IV × 1 then 1

mg/kg IV

after HD

CRRT: 2

mg/kg IV × 1

then 1.5

mg/kg IV

every 24

hours

TMP/SMX

IVPO

When switching

to oral therapy,

consider that a

single-strength

tablet has 80 mg

of TMP, a

double-strength

tablet 160 mg of

TMP.

Systemic GNR infections

10 mg TMP/kg/day IV

divided every 6–12 hours

Pneumocystis pneumonia

15–20 mg TMP/kg/day IV

divided every 6–8 hours

5–7.5 mg TMP/kg/day IV

divided every 12–24 hours

10–15 mg TMP/kg/day IV

divided every 12–24 hours

2.5–5.0 mg

TMP/kg IV

every 24

hours

5–10 mg

TMP/kg IV

every 24

hours

HD: 2.5–5

mg

TMP/kg/day

every 24

hours

CRRT: 5–

7.5mg

TMP/kg/day

divided every

12–24 hours

Vancomycin

Uncomplicated

infections

Serious

infections

>60

mL/minute

10–15 mg/kg

IV every 12

hours

1

15–20 mg/kg

IV every 8–12

hours

2

40–60

mL/minute

10–15 mg/kg

IV every 12–

24 hours

20–40

mL/minute

5–10 mg/kg

IV every 24

hours

10–20

mL/minute

5–10 mg/kg

IV every 24–

48 hours

<10

mL/minute

10–15 mg/kg

IV loading

dose ×1;

redose

according to

serum levels

HD: 15–20

mg/kg load,

then 500 mg

IV after HD

only

CRRT: 10–15

mg/kg IV

every 24

hours

Round dose to 250 mg, 500 mg, 750 mg, 1 g, 1.25 g, 1.5 g, 1.75 g, or 2 g (maximum 2 g/dose). Trough levels

should be obtained within 30 minutes before fourth dose of a new regimen or dosage change. Vancomycin

troughs are not recommended in patients in whom anticipated duration of therapy is ≤3 days.

1 For patients with uncomplicated infections requiring vancomycin, trough levels of 10–15 mcg/mL are

recommended.

2 For patients with serious infections caused by MRSA (central nervous system infections, endocarditis,

ventilator-associated pneumonia, bacteremia, or osteomyelitis), trough levels of 15–20 mcg/mL are

recommended.

Voriconazole

IVPO

LD = 400 mg every 12 hours

× 1 day, then 200 mg every 12

hours (PO)

No change No change* No change*

PO should be used when possible, as oral bioavailability >95%. IV dose: LD = 6 mg/kg/dose every 12 hours × 1

day, then 4 mg/kg/dose every 12 hours.

*The use of the IV formulation should be avoided in patients with CrCl <50 mL/minute owing to accumulation of

IV vehicle and is contraindicated in ESRD and hemodialysis. May require dose adjustment in hepatic

dysfunction.

Doses are those recommended for systemic infections in hospitalized patients commonly treated with these agents

for moderate/severe infections. Abstracted in part from doses developed by the UCSF Antibiotic Advisory

Subcommittee and the Pharmacy & Therapeutics Committee (updated 6/2015). More mild infection may require

decreased doses compared with moderate to severe infection. Dosing guidelines may differ in other institutions.

Estimate of renal function using Cockcroft and Gault equation: CrCl (mL/minute) = (140 – age) × Wt (kg)/72 ×

SCr (mg/dL) (for females multiply by 0.85). Ideal body weight equation:

Males: IBW = 50 kg + 2.3 kg for each inch over 5 feet.

Females: IBW = 45.5 kg + 2.3 kg for each inch over 5 feet.

Adjusted body weight: ABW = IBW + 0.4 (actual weight - IBW).

CrCl, creatinine clearance; CRRT, continuous renal replacement therapy (assumes an ultrafiltration rate of 2

L/hour with continuous venovenous hemofiltration and an ultrafiltration rate of 1 L/hour and dialysate flow rate of

1 L/hour with continuous venovenous hemodiafiltration and residual native glomerular filtration rate <10

mL/minute); ESRD, end-stage renal disease; HD, intermittent (high-flux) hemodialysis (when administering a daily

dose with HD, the drug should be administered after the HD session); HSV, herpes simplex virus; IV, intravenous;

IV-PO, high oral bioavailability (consider initiating with or switching to PO therapy when patient tolerating orals);

LD, loading dose; MRSA, methicillin-resistant Staphylococcus aureus; PO, by mouth; SCr, serum creatinine.

p. 1339

p. 1340

Although renal function can be approximated with the use of the Cockcroft and

Gault equation (or a similar equation), hepatic function is more difficult to evaluate.

No standard liver function test (AST, ALT, alkaline phosphatase) has been

demonstrated to correlate well with hepatic drug clearance. Some tests, such as PT,

INR, and albumin, are markers of hepatic function, but even these tests do not clearly

predict drug clearance. Patients receiving hemodialysis or continuous hemofiltration

provide additional dosing challenges. Table 62-9 provides dosing recommendations

in patients receiving hemodialysis or continuous hemofiltration.

Patient Age

It is important to note that most dosing information is derived from a younger,

relatively healthy patient population. It is clear that the very young and the elderly

have a decreased ability to clear drugs; thus, dosage requirements for many agents

are likely to be decreased in neonatal and geriatric patients.

Fever and Inoculum Effect

The impact of other factors on the selection of an antimicrobial dose is less clear.

Fever increases and decreases blood flow to mesenteric, hepatic, and renal organ

systems,

29 and it can either increase or decrease drug clearance. Inoculum effect has

taken place when higher concentrations of a bacterial inoculum result in an increase

in the MIC.

30 As an example, piperacillin may demonstrate an MIC of 8.0 mcg/mL

against P. aeruginosa at a concentration of 10

5 colony-forming units/mL (CFU/mL);

however, at 10

9 CFU/mL, the MIC may increase to 32 to 64 mcg/mL. This

phenomenon is well recognized, particularly with β-lactamase–producing bacteria

treated with β-lactam antimicrobials. The more stable the antimicrobial is to βlactamase, the less the influence of the inoculum effect. Aminoglycosides,

quinolones, and imipenem appear to be less affected by the inoculum effect than βlactams. The inoculum effect probably is most relevant in the treatment of a bacterial

abscess, in which extremely high concentrations of bacteria would be expected. As a

result, antimicrobials that are more susceptible to the inoculum effect may require

increased drug dosages for optimal outcome in the treatment of abscesses.

PHARMACOKINETICS AND

PHARMACODYNAMICS

CASE 62-1, QUESTION 12: R.G.’s respiratory status remains unchanged; thus, the ciprofloxacin is

discontinued and cefepime and gentamicin are started empirically. The use of a prolonged 3 hour IV infusion of

cefepime is being considered in R.G. In addition, the use of single daily dosing of gentamicin is being discussed.

What is the rationale for these approaches, and would either be advantageous for R.G.?

β-Lactams, such as cefepime, are not associated with increased bacterial killing

with increasing drug concentrations. Pharmacodynamic activity with β-lactams best

correlates with the duration of time that antimicrobial levels are maintained above

the MIC.

31 The animal model suggests that β-lactam antimicrobials should be dosed

such that their serum levels exceed the MIC of the pathogen as long as possible.

31

This observation appears to be most important in the neutropenic model, in which the

use of a constant infusion more reliably inhibits bacterial growth compared with

traditional intermittent dosing. An additional benefit of the use of constant infusions

of β-lactams is that smaller daily doses appear to be as effective as higher doses

administered intermittently. Other than this latter outcome, it is unclear, however,

whether constant infusions have any distinct advantages or disadvantages compared

with usual dosing of β-lactams. The efficacy of quinolones, vancomycin, and

daptomycin best correlates with the peak plasma concentration to MIC ratio or area

under the curve (AUC) to MIC ratio.

31

In light of this pharmacodynamic principle, it

is possible that ciprofloxacin was underdosed in this patient, contributing to the

therapeutic failure, particularly if the MIC was in the upper range of susceptibility

for this agent.

Aminoglycosides traditionally have been administered every 8 to 12 hours to

achieve peak serum gentamicin levels of 5 to 8 mcg/mL to ensure efficacy in the

treatment of serious gram-negative infection.

32

,

33 Gentamicin troughs of greater than 2

mcg/mL have been associated with an increased risk for nephrotoxicity.

33

,

34 These

studies attempting to correlate efficacy and toxicity with serum levels and the

association of peaks or troughs with clinical outcomes have been questioned.

21

Vancomycin troughs of 5 to 10 mcg/mL have been traditionally recommended

35

,

36

;

however, current recommendations suggest higher troughs (10–20 mcg/mL)

depending on the pathogen, site of infection, and severity of illness.

37

Several antimicrobials (e.g., aminoglycosides) have been associated with a

pharmacodynamic phenomenon known as a post-antibiotic effect (PAE). PAE is

delayed regrowth of bacteria after exposure to an antibiotic

31

,

38

(i.e., continued

suppression of normal growth in the absence of antibiotic levels above the MIC of

the organism). As an example, if P. aeruginosa is cultured in broth, it will multiply to

a concentration of 10

9 CFU/mL. If piperacillin is added in a concentration above the

MIC for the organism, a reduction in the bacterial concentration is observed. As

described previously, a β-lactam antibiotic should be present in concentrations

above the MIC to optimize its time-dependent killing. When piperacillin is removed

from the broth, immediate bacterial growth takes place. If the above experiment is

repeated with gentamicin, a reduction in bacterial CFU is observed. In contrast to

that observed with β-lactam antibiotics, if the gentamicin is removed from the system,

a lag period of 2 to 6 hours takes place before characteristic bacterial growth occurs.

This lag period is defined as the PAE. A PAE has also been observed with

quinolones and imipenem against gram-negative bacteria. Although most β-lactam

antibiotics, such as antipseudomonal penicillins or cephalosporins, do not exhibit

PAE with gram-negative organisms, PAE has been demonstrated with β-lactam with

gram-positive pathogens such as S. aureus.

Once-Daily Dosing of Aminoglycosides

As a result of PAE and other pharmacodynamic factors, certain antimicrobials may

be dosed less frequently. The greatest clinical experience has been with the

aminoglycosides in the treatment of gram-negative infection.

39

,

40 Earlier data

suggested that the maximal aminoglycoside peak level to MIC ratio correlates well

with clinical response. Thus, the higher the achievable peak, the greater likelihood of

a favorable outcome. Consequently, greater, less frequent doses of aminoglycosides

should work at least as well as the more traditional lower, more frequent doses.

Once-daily dosing of aminoglycosides in the treatment of gram-negative infection is

as efficacious as traditional multiple daily dosing.

21

Single daily dosing of aminoglycosides has been investigated primarily in patients

with normal renal function, and few critically ill patients have been treated with this

nontraditional regimen.

p. 1340

p. 1341

Thus, patients in septic shock are less clear candidates for once-daily dosing. The

utility and proper timing of serum aminoglycoside concentrations and association

with clinical outcomes are debatable with nontraditional once-daily

aminoglycosides.

In summary, the use of a prolonged IV infusion of cefepime is possible in R.G., but

the benefit of this mode of administration is not clear. Considering the severity of

R.G.’s infection and his elevated serum creatinine level, he is not a candidate for

single daily dosing of aminoglycosides (i.e., 5–6 mg/kg every 24 hours). Independent

of the aminoglycoside-associated PAE, his current renal function requires a reduced

gentamicin dose to treat his infection.

Antimicrobial Protein Binding

CASE 62-1, QUESTION 13: Ceftriaxone (Rocephin), rather than cefepime, is being considered for the

treatment of R.G.’s infection. Ceftriaxone is more highly protein bound than cefepime. Why is protein binding

important in the selection of therapy?

Free (i.e., unbound) rather than total drug levels are best correlated with

antimicrobial activity,

41 and the degree of protein binding may have important

clinical consequences in some patients. Chambers et al.

42

reported treatment failures

with the highly protein-bound cefonicid (98% protein bound) in patients with

endocarditis caused by S. aureus. Despite achievable serum drug concentrations well

above the MIC of the organism, breakthrough bacteremia occurred in three of four

patients. Although total drug concentrations greatly exceeded the MIC of the

pathogen, free concentrations were consistently below the level necessary to inhibit

bacterial growth. Similar experiences have been reported with daptomycin (90% to

93% protein bound).

43 Thus, clinical cure appears to be more likely if unbound

antibiotic concentrations exceed the MIC of the infecting organism. Although

ceftriaxone is 85% to 90% protein bound, the free concentrations probably remain

far above the MIC of the Serratia. Therefore, protein-binding considerations are

unlikely to be important in the treatment of R.G.’s infection.

ANTIMICROBIAL FAILURE

Antibiotic-Specific Factors

CASE 62-1, QUESTION 14: Despite “appropriate” treatment, R.G. is unresponsive to antimicrobial therapy.

What antibiotic-specific factors may contribute to “antimicrobial failure”?

Antimicrobials may fail for various reasons, including patient-specific host

factors, drug or dosage selection, and concomitant disease states. One of the most

common reasons for antimicrobial failure is drug resistance.

44–46 Several clinically

important pathogens have been associated with emergence of resistance during the

past decade, including M. tuberculosis,

47 enterococci,

48 gram-negative rods,

44 S.

aureus,

49 S. pneumoniae,

50 and others. Of particular concern is the isolation of

glycopeptide-resistant S. aureus,

49 and multidrug-resistant Acinetobacter and

Pseudomonas.

51 Development of resistance during therapy, although less common

than initial intrinsic resistance, may also account for failure to respond to therapy.

Organisms that produce extended-spectrum β-lactamase or amp C β-lactamases may

be unresponsive to β-lactam therapy despite associated in-vitro susceptibility.

52

Superinfection may also play a role in the unsuccessful treatment of infection.

Superinfection has taken place when a new pathogen resistant to the current

antimicrobial regimen is isolated. If R.G.’s ceftriaxone-treated Serratia pneumonia

subsequently worsens and a tracheal aspirate returns positive for P. aeruginosa, then

supercolonization and, perhaps, superinfection have occurred.

Combination Therapy

Most infections can be treated with monotherapy (e.g., an E. coli wound infection is

treatable with a cephalosporin). Some infections, however, require two-drug therapy,

including most cases of enterococcal endocarditis and perhaps certain P. aeruginosa

infections. Hilf et al.

53 studied 200 consecutive patients with P. aeruginosa

bacteremia and demonstrated a 47% mortality in those receiving monotherapy

(antipseudomonal β-lactam or aminoglycoside) versus 27% in those in whom twodrug therapy was used. Thus, monotherapy appeared to contribute to antimicrobial

failure in this specific study.

In contrast to the findings of the previous trial, almost all later investigations do

not support the use of two drugs over monotherapy in the treatment of serious gramnegative infection, including P. aeruginosa.

54–56 An exception to this rule is

bacteremia caused by P. aeruginosa in neutropenic patients.

If two antimicrobials are used in the treatment of infection, one of three sequelae

will result: indifference, synergism, or antagonism.

57

Indifference occurs when the

antimicrobial effect of drug A plus that of drug B equals the anticipated sum activity

of the two drugs. Although numerous definitions exist, synergism generally occurs

when the addition of drug A to drug B results in a total antibiotic activity greater than

the expected sum of the two agents. Antagonism occurs if the addition of drug A to

drug B results in a combined activity less than the sum of drug A plus drug B. An

example of antagonism is the combination of imipenem with a less β-lactamase–

stable β-lactam, such as piperacillin.

58

If P. aeruginosa is exposed to imipenem and

piperacillin, the imipenem induces the organism to produce increased β-lactamase.

Imipenem is remarkably β-lactamase stable and is not degraded by this β-lactamase.

In direct contrast, piperacillin is easily degraded by the β-lactamase. Thus, imipenem

antagonized the effectiveness of piperacillin. Antagonism is not unique to

antibacterials; itraconazole may antagonize amphotericin B in the treatment of certain

fungal infections.

59

Pharmacologic Factors

CASE 62-1, QUESTION 15: What pharmacologic or pharmaceutic factors may be implicated in failure of

therapy?

Subtherapeutic dosing regimens are commonplace, particularly for agents with a

low therapeutic index, such as the aminoglycosides. For example, a serious gramnegative pneumonia may not respond to aminoglycoside therapy if the achievable

peak gentamicin serum levels are only 3 to 4 mcg/mL.

21

,

32 Considering that only 20%

to 30% of the aminoglycoside penetrates from serum into bronchial secretions, only

0.5 to 1.0 mcg/mL may exist at the site of infection,

60 a level that may be inadequate

to treat pneumonia. Another example of dosing contributing to antimicrobial failure

centers on the use of loading doses. Aminoglycosides or vancomycin should be

initiated with a loading dose, particularly in patients with renal failure. If the

clinician neglects to use a loading dose, it may take several days before a therapeutic

level is achieved. As described previously, yet another reason for subtherapeutic

antimicrobial levels and potential drug failure is reduced oral absorption secondary

to drug interactions (e.g., concomitant oral ciprofloxacin with antacids or iron).

p. 1341

p. 1342

An emerging problem relates to the use of vancomycin in the treatment of serious

methicillin-resistant S. aureus (MRSA) infection. By CLSI standards, an isolate of

MRSA with an MIC of 2 mcg/mL is considered susceptible. Current vancomycin

dosing schemes are designed to achieve an AUC/MIC ratio of ≥400 to ensure

maximal efficacy. However, a meta-analysis of patients with S. aureus bacteremia

demonstrated no differences in the risk of death when comparing patients with S.

aureus exhibiting high-vancomycin MIC (≥1.5 mg/L) to those with low-vancomycin

MIC (<1.5 mg/L).

61 The infection site also potentially contributes to antimicrobial

failure. Most antimicrobials concentrate in the urine, resulting in therapeutic levels

even with low doses. In some infections, such as meningitis, prostatitis, and

endophthalmitis, antimicrobial penetration to the site of infection may be inadequate.

Agents that penetrate well into these sites are associated with a more favorable

outcome.

Another potential reason for antimicrobial failure is inadequate therapy duration.

A woman with a first-time uncomplicated cystitis may respond adequately to a 3-day

course of an antibiotic. In contrast, patients with recurrent urinary tract infections are

not candidates for this short course of therapy, however, and failure would be

expected with only 3 days of therapy.

Host Factors

CASE 62-1, QUESTION 16: What host factors may contribute to the failure of antimicrobial therapy?

Several host factors may limit the ability of an antibiotic to cure infection.

Infection of prosthetic material (e.g., IV catheters, orthopedic prostheses, mechanical

cardiac valves, and vascular grafts) is difficult to eradicate without removal of the

hardware. In most cases, surgical intervention is necessary. To treat R.G.’s IV

catheter infection adequately, removal of his central intravenous catheter would be

optimal. Similar to removal of prostheses, large undrained abscesses are difficult, if

not impossible, to treat with antimicrobial therapy. These infections generally require

surgical drainage for successful outcome.

Diabetic foot ulcer cellulitis may not respond adequately to antimicrobial therapy.

Reasons for antimicrobial failure in patients with diabetes include poor wound

healing and reduced delivery of antibiotics to the infection site.

Immune status, particularly neutropenia or lymphocytopenia, also affects the

outcome in the treatment of infection. Profoundly, neutropenic patients with

disseminated Aspergillus infections are unlikely to respond to even the most

appropriate antifungal therapy. Similarly, patients with AIDS, who have low CD4

lymphocyte counts, cannot eradicate various infections, including those caused by

cytomegalovirus, atypical mycobacteria, and cryptococci.

Once these factors have been eliminated as causes for antimicrobial failure,

noninfectious sources must be ruled out. As discussed, malignancy, autoimmune

disease, drug fever, and other diseases must be evaluated.

CASE 62-1, QUESTION 17: Other than initiation of adequate antimicrobial therapy, what adjunct measures

can be considered in this patient with septic shock?

The 2013 Surviving Sepsis Campaign: International Guidelines for Management of

Severe Sepsis and Septic Shock consultants developed key recommendations toward

the early goal-directed resuscitation of the septic patient.

62 These recommendations

include the use of “sepsis bundles”, i.e., multiple interventions taking place at the

same time. Key recommended adjuncts include administration of broad-spectrum

antibiotics within 1 hour of diagnosis of septic shock, administration of either

crystalloid or colloid fluid resuscitation, and norepinephrine or dopamine to maintain

mean arterial pressure of at least 65 mm Hg. In addition, stress-dose steroid therapy

can be given to those patients whose blood pressure is poorly responsive to fluid

resuscitation and vasopressors. While one might expect improved survival with

adherence to such guidelines, the results have been mixed. A large meta-analysis

evaluated the influence of performance improvement programs regarding compliance

with sepsis bundles. These programs have been found to be associated with

increased adherence to resuscitation and management sepsis bundles and with

reduced mortality in patients with sepsis, severe sepsis, or septic shock.

63

In contrast,

in patients with septic shock who were identified early and received intravenous

antibiotics and adequate fluid resuscitation, hemodynamic management according to a

strict early goal-directed therapy protocol did not lead to an improvement in

outcome.

64

KEY REFERENCES AND WEBSITES

A full list of references for this chapter can be found at

http://thepoint.lww.com/AT11e. Below are the key references and websites for this

chapter, with the corresponding reference number in this chapter found in parentheses

after the reference.

Key References

Angus DC, van der Poll T. Severe sepsis and septic shock. N EnglJ Med. 2013;369(9):840. (3)

Boucher HW et al. Bad bugs, no drugs: No ESKAPE! An update from the Infectious Diseases Society of

America. Clin Infect Dis. 2009;48(1):1. (44)

Czock D et al. Pharmacokinetics and pharmacodynamics of antimicrobial drugs. Expert Opin Drug Metab Toxicol.

2009;5(5):475. (31)

Dellinger RP et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic

shock. Intensive Care Med. 2013;39(2):165. (62)

Hooton TM et al. Diagnosis, prevention, and treatment of catheter-associated urinary tract infection in adults: 2009

International Clinical Practice Guidelines from the Infectious Diseases Society of America. Clin Infect Dis.

2010;50(5):625. (12)

Horowitz HW. Fever of unknown origin or fever of too many origins? N EnglJ Med. 2013;368(3):197. (10)

Kalil AC et al. Association between vancomycin minimum inhibitory concentration and mortality among patients

with Staphylococcus aureus bloodstream infections: a systematic review and meta-analysis. JAMA.

2014;312(15):1552. (61)

Mandell LA et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the

management of community-acquired pneumonia in adults. Clin Infect Dis. 2007;44(Suppl 2):S27. (15)

Rybak MJ et al. Vancomycin therapeutic guidelines: a summary of consensus recommendations from the

infectious diseases Society of America, the American Society of Health-System Pharmacists, and the Society of

Infectious Diseases Pharmacists. Clin Infect Dis. 2009;49(3):325. (37)

Solomkin JS et al. Diagnosis and management of complicated intra-abdominal infection in adults and children:

guidelines by the Surgical Infection Society and the Infectious Diseases Society of America [published

correction appears in Clin Infect Dis. 2010;50(12):1695]. Clin Infect Dis. 2010;50(2):133. (13)

Vincent JL et al. Evolving concepts in sepsis definitions. Crit Care Clin. 2009;25(4):665, vii. (1)

Wacker C et al. Procalcitonin as a diagnostic marker for sepsis: a systematic review and meta-analysis. Lancet

Infect Dis. 2013;13(5):426. (4)

Weisfelt M et al. Bacterial meningitis: a review of effective pharmacotherapy. Expert Opin Pharmacother.

2007;8(10):1493. (25)

COMPLETE REFERENCES CHAPTER 62 PRINCIPLES OF

INFECTIOUS DISEASES

Vincent JL et al. Evolving concepts in sepsis definitions. Crit Care Clin. 2009;25(4):665, vii.

Lee WL, Slutsky AS. Sepsis and endothelial permeability. N EnglJ Med. 2010;363(7):689.

Angus DC, van der Poll T. Severe sepsis and septic shock. N EnglJ Med. 2013;369(9):840.

Wacker C et al. Procalcitonin as a diagnostic marker for sepsis: a systematic review and meta-analysis. Lancet

Infect Dis. 2013;13(5):426.

Phua J et al. Has mortality from acute respiratory distress syndrome decreased over time? A systematic review.

Am J Respir Crit Care Med. 2009;179(3):220.

Zambon M, Vincent JL. Mortality rates for patients with acute lung injury/ARDS have decreased over time.

Chest. 2008;133(5):1120.

Zarrouk V et al. Evaluation of the management of postoperative aseptic meningitis. Clin Infect Dis.

2007;44(12):1555.

Lee BE, Davies HD. Aseptic meningitis. Curr Opin Infect Dis. 2007;20(3):272.

Kayoko H et al. Fever of unknown origin: an evidence-based review. Am J Med Sci. 2012;344(4):307.

Horowitz HW. Fever of unknown origin or fever of too many origins? N EnglJ Med. 2013;368(3):197.

Patel RA, Gallagher JC. Drug fever. Pharmacotherapy. 2010;30(1):57.

Hooton TM et al. Diagnosis, prevention, and treatment of catheter-associated urinary tract infection in adults:

2009 International Clinical Practice Guidelines from the Infectious Diseases Society of America. Clin Infect

Dis. 2010;50(5):625.

Solomkin JS et al. Diagnosis and management of complicated intra-abdominal infection in adults and children:

guidelines by the Surgical Infection Society and the Infectious Diseases Society of America [published

correction appears in Clin Infect Dis. 2010;50(12):1695]. Clin Infect Dis. 2010;50(2):133.

Chalmers JD et al. Healthcare-associated pneumonia does not accurately identify potentially resistant pathogens:

a systematic review and meta-analysis. Clin Infect Dis. 2014;58(3):330

Mandell LA et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on

the management of community-acquired pneumonia in adults. Clin Infect Dis. 2007;44(Suppl 2):S27.

Bennett JE et al, eds. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 8th ed.

Philadelphia, PA: Elsevier Saunders; 2015.

Clinical and Laboratory Standards Institute (CLSI). Performance Standards for Antimicrobial Susceptibility

Testing; Twenty-fifth Informational Supplement. CLSI Document M100-S25. Wayne, PA: CLSI; 2015.

Scheutz AN. Antimicrobial resistance and susceptibility testing of anaerobic bacteria. Clin Infect Dis.

2014;59(5):698.

Cantón E et al. Trends in antifungalsusceptibility testing using CLSI reference and commercial methods. Expert

Rev Anti Infect Ther. 2009;7(1):107.

Pfaller MA et al. Wild-type MIC distribution and epidemiological cutoff values for Aspergillus fumigatus and

three triazoles as determined by the Clinical and Laboratory Standards Institute broth microdilution methods. J

Clin Microbiol. 2009;47(10):3142.

Drusano GL et al. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect

Dis. 2007;45(6):753.

Cosgrove SE. The relationship between antimicrobial resistance and patient outcomes: mortality, length of hospital

stay, and health care costs. Clin Infect Dis. 2006;42(Suppl 2): S82.

Bhavnani SM et al. Cost-effectiveness of daptomycin versus vancomycin and gentamicin for patients with

methicillin resistant Staphylococcus aureus bacteremia and/or endocarditis. Clin Infect Dis. 2009;49(5):691.

Spellberg B et al. The epidemic of antibiotic-resistant infections: a call to action for the medical community from

the Infectious Diseases Society of America. Clin Infect Dis. 2008;46(2):155.

Weisfelt M et al. Bacterial meningitis: a review of effective pharmacotherapy. Expert Opin Pharmacother.

2007;8(10):1493.

López-Cabezas C et al. Antibiotics in endophthalmitis: microbiological and pharmacokinetic considerations. Curr

Clin Pharmacol. 2010;5(1):47.

Lipsky BA et al. Treatment of bacterial prostatitis. Clin Infect Dis. 2010;50(12):1641.

Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31.

Mackowiak PA. Influence of fever on pharmacokinetics. Rev Infect Dis. 1989;11(5):804.

Brook I. Inoculum effect. Rev Infect Dis. 1989;11(3):361.

Czock D et al. Pharmacokinetics and pharmacodynamics of antimicrobial drugs. Expert Opin Drug Metab

Toxicol. 2009;5(5):475.

Moore RD et al. Association of aminoglycoside levels with therapeutic outcome in gram-negative pneumonia. Am

J Med. 1984;77(4):657.

Mattie H et al. Determinants of efficacy and toxicity of aminoglycosides. J Antimicrob Chemother.

1989;24(3):281.

Matske GR et al. Controlled comparison of gentamicin and tobramycin nephrotoxicity. Am J Nephrol.

1983;3(1):11.

Begg EG et al. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharmacol. 2001;52(Suppl 1):35S.

MacGowan AP. Pharmacodynamics, pharmacokinetics, and therapeutic drug monitoring of glycopeptides. Ther

Drug Monit. 1998;20(5):473.

Rybak MJ et al. Vancomycin therapeutic guidelines: a summary of consensus recommendations from the

infectious diseases Society of America, the American Society of Health-System Pharmacists, and the Society

of Infectious Diseases Pharmacists. Clin Infect Dis. 2009;49(3):325.

Pea F, Viale P. The antimicrobial therapy puzzle: could pharmacokinetic-pharmacodynamic relationships be

helpful in addressing the issue of appropriate pneumonia treatment in critically ill patients? Clin Infect Dis.

2006;42(12):1764.

Hatala R, Dinh TT, Cook DJ. Single daily dosing of aminoglycosides in immunocompromised adults: a systematic

review. Clin Infect Dis. 1997;24(5):810.

Ferriols-Lisart R, Alos-Alminana M. Effectiveness and safety of once-daily aminoglycosides: a meta-analysis.

Am J Health Syst Pharm. 1996;53(10):1141.

Schmidt S et al. Effect of protein binding on the pharmacological activity of highly bound antibiotics. Antimicrob

Agents Chemother. 2008;52(11):3994.

Chambers HF et al. Failure of a once-daily regimen of cefonicid for treatment of endocarditis due to

Staphylococcus aureus. Rev Infect Dis. 1984;6(Suppl 4):S870.

Schwartz BS et al. Daptomycin treatment failure for vancomycin-resistant Enterococcus faecium infective

endocarditis: impact of protein binding? Ann Pharmacother. 2008;42(2):289.

Boucher HW et al. Bad bugs, no drugs: No ESKAPE! An update from the Infectious Diseases Society of

America. Clin Infect Dis. 2009;48(1):1.

Giamarellou H, Poulakou G. Multidrug-resistant gram negative infections: what are the treatment options? Drugs.

2009;69(14):1879.

Qureshi ZA et al. Colistin-resistant Acinetobacter baumanii: beyond carbapenem resistance. Clin Infect Dis.

2015;60(9):1295.

Jassal M, Bishai WR. Extensively drug-resistant tuberculosis. Lancet Infect Dis. 2009;9(1):19.

Reik R et al. The burden of vancomycin-resistant enterococcal infections in US hospitals, 2003 to 2004. Diagn

Microbiol Infect Dis. 2008;62(1):81.

Sievert DM et al. Vancomycin-resistant Staphylococcus aureus in the United States, 2002–2006. Clin Infect Dis.

2008;46(5):668.

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more