14.01.2015 Views

Pharmacotherapy Review Packet

Pharmacotherapy Review Packet

Pharmacotherapy Review Packet

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Anthelmintics<br />

AHFS 080800<br />

October 27, 2004<br />

I. Overview<br />

In the United States, helminthic infections are most frequently seen in recent immigrants from<br />

Southeast Asia, the Caribbean, Mexico, and Central America. 1 There is a higher incidence of<br />

helminthic infections in the southern states. Other populations that have a high risk of infestation<br />

include institutionalized patients (both young and elderly), preschool children in day care centers,<br />

residents of Native American reservations, and homosexual individuals. Certain conditions and<br />

drugs (fever, corticosteroids, and anesthesia) can cause atypical localization of worms.<br />

Additionally, immunocompromised hosts can be overwhelmed by some helminthic infections,<br />

such as strongyloidiasis.<br />

The majority of intestinal helminthic infections are not associated with clearly defined<br />

manifestation of disease, but they can cause significant pathology. One factor that determines the<br />

pathogenicity of helminthes is their population density. Light infections may be fairly well<br />

tolerated, whereas high populations of intestinal helminthes can result in predictable disease<br />

presentations. Significant advances have occurred in the past 20 years in the drugs available to<br />

treat parasitic helminthes. Single-dose treatments are available for most of the common species of<br />

worms. After reviewing clinical data, the World Health Organization had endorsed concomitant<br />

use of praziquantel and albendazole to treat intestinal worms and schistosomiasis. 2<br />

There are two main types of helminthic infections, those caused by flatworms (tapeworms and<br />

flukes) and those caused by roundworms (pinworm, whipworm, ascaris, and strongyloides).<br />

Helminthic diseases are caused by nematodes and include hookworm disease, ascariasis and<br />

enterobiasis. Morbidity and disease with intestinal nematodes is related to the intensity of<br />

infection or worm burden; subjects with transient exposure have less-severe disease. Adverse<br />

events of intestinal nematodes are malnutrition, fatigue, and diminished work capacity. Treatment<br />

with anthelmintic agents results in complete eradication and significant change in well-being.<br />

Hookworm<br />

Hookworm is caused by an infection of the small intestine caused by either Ancylostoma<br />

duodenale or Necator americanus. N. americanus is found in the southeastern U.S., where the<br />

temperature and humidity provide the proper envorinment. 1 Ancylostoma is rare in the U.S. The<br />

life cycles of both species of hook worm are similar. The adult worms live in the small intestine<br />

attached to the mucosa. The females liberate eggs, which are eliminated in the feces and develop<br />

into larvae. Infective larva enter the host in contaminated food or water, or penetrate the skin,<br />

where a papular eruption with localized edema and erythema can result. Injury to the small<br />

intestine can cause mechanical and lytic destruction of tissue and the loss of blood can lead to<br />

anemia and hypoproteinemia. Eosinophilia (30-60%) is present in patients with chronic infection.<br />

Stool should be examined for eggs and larvae.<br />

Ascariasis<br />

This infection is caused by the giant roundworm Ascaris lumbricoides. Female worms can range<br />

from 20-35cm in length. The worm is found worldwide, but more commonly in areas where<br />

sanitation is poor. 1 In the United States., endemic areas include southeastern parts of the<br />

Appalachian and the Gulf coast states. Estimates place nearly 4 million people in the U.S. with<br />

ascariasis. Clinical manifestations of this disease include pneumonitis, fever, cough, eosinophilia,<br />

and pulmonary infiltrates, as the larvae migrate through the lungs. Diagnosis is made by<br />

demonstrating the characteristic egg in the stool.<br />

1


Enterobiasis<br />

Also known as pinworm infection, is caused by Enterobius vermicularis. The pinworm is a small<br />

thread-like spindle-shaped worm about cm in length. It is the most widely distributed helminthic<br />

infection in the world. There are estimates of 42 million cases in the United States, a majority of<br />

which are children. The most common problem with infection is cutaneous irritation in the<br />

perianal region, made by the migrating females or presence of eggs. Scratching can lead to<br />

dermatitis and secondary bacterial infections.<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this review.<br />

Table 1. Anthelmintics in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name Rx vs. OTC<br />

Albendazole Oral Tablet Albenza Rx<br />

Ivermectin Oral Tablet Stromectol Rx<br />

Mebendazole Oral Chewable Tablets *Vermox Rx<br />

Praziquantel Oral Tablet Biltricide Rx<br />

Pyrantel Pamoate Oral Liquid/Soft Gel Capsules Antiminth (D/c’d 11-2003), Pin-X, OTC<br />

Reeses’s Pinworm Medicine, Pinworm<br />

Treatment<br />

Thiabendazole Oral Suspension, Chewable Tablets Mintezol Rx<br />

*Generic Available.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

The Centers for Disease Control and Prevention have instituted recommendations for preventing<br />

opportunistic infections among hematopoietic stem cell transplant (HSCT) recipients, and includes<br />

prevention of helminthic infections. 3 Recommendations include treatments for Strongyloides<br />

stercoralis. Recipients should avoid contact with out houses and cutaneous exposure to soil or<br />

other surfaces that might be contaminated with human feces. All recipients who work in the<br />

health care industry (hospitals or institutions) where they might be exposed to fecal matter, should<br />

wear gloves.<br />

Travel and residence histories should be obtained for all patients before HSCT to determine any<br />

exposures to high-risk areas (e.g., such moist temperate areas as the tropics, subtropics, or the<br />

southeastern United States and Europe). 3 HSCT candidates who have unexplained peripheral<br />

eosinophilia or who have resided in or traveled to areas endemic for strongyloidiasis, even during<br />

the distant past, should be screened for asymptomatic strongyloidiasis before HSCT. Serologic<br />

testing with an enzyme-linked immunosorbent assay is the preferred screening method and has a<br />

sensitivity and specificity of >90%. FDA-licensed or -approved screening tests should be used.<br />

Although stool examinations for strongyloidiasis are specific, the sensitivity obtained from >3<br />

stool examinations is 60%-70%; the sensitivity obtained from concentrated stool exams is, at best,<br />

80%. A total of >3 stool examinations should be performed if serologic tests are unavailable or if<br />

strongyloidiasis is clinically suspected in a seronegative patient.<br />

HSCT candidates whose screening tests before HSCT are positive for Strongyloides species, and<br />

those with an unexplained eosinophilia and a travel or residence history indicative of exposure to<br />

Strongyloides stercoralis should be empirically treated before transplantation, preferably with<br />

ivermectin, even if seronegative or stool-negative.<br />

2


To prevent recurrence among HSCT candidates with parasitologically confirmed strongyloidiasis,<br />

cure after therapy should be verified with >3 consecutive negative stool examinations before<br />

proceeding with HSCT. Data are insufficient to recommend a drug prophylaxis regimen after<br />

HSCT to prevent recurrence of strongyloidiasis. HSCT recipients who had strongyloidiasis before<br />

or after HSCT should be monitored carefully for signs and symptoms of recurrent infection for 6<br />

months after treatment. Hyperinfection strongyloidiasis has not been reported after autologous<br />

HSCT; however, the same screening precautions should be used among autologous recipients.<br />

Indications for empiric treatment for strongyloidiasis before HSCT are the same among children<br />

or adults except for children weighing


III.<br />

Comparative Indications of the Anthelmintic Antibiotics<br />

Table 2 lists the FDA-approved indications for the anthelmintic antibiotics. Mebendazole is indicated for use as described in Table 2, and is also indicated<br />

for single or mixed infections. Table 3 further describes the major intestinal parasites and the treatments of choice.<br />

Table 2. FDA-Approved Indications for the Anthelmintics 5-7<br />

Drug Neurocysticercosis<br />

Hydatid<br />

Disease<br />

Strongyloidiasis<br />

Onchocerciasis<br />

Pinworm<br />

Albendazole<br />

✔ ✔ *<br />

Whipworm<br />

(Trichuris<br />

richiura)<br />

Roundworm<br />

(Ascaris)<br />

Hookworm<br />

(Common<br />

and<br />

American)<br />

All species<br />

of<br />

Schistosoma<br />

Liver<br />

Flukes<br />

Clonorchis<br />

sinensis /<br />

Opisthorchis<br />

viverrini<br />

Cutaneous<br />

Larva<br />

Migrans<br />

(creeping<br />

eruption)<br />

Visceral<br />

Larva<br />

Migrans<br />

Trichinosis<br />

Ivermectin<br />

✔ ✔<br />

Mebendazole<br />

✔ ✔ ✔ ✔<br />

Praziquantel<br />

✔ ✔ ✔<br />

Pyrantel<br />

Pamoate<br />

✔ ✔<br />

Thiabendazole<br />

✔ ✔ ** ✔ ■ ✔ ■ ✔ ■ ✔ ✔ ✔<br />

* Disease of the liver, lung, and peritoneum caused by the larval form of the dog tapworm, Echinococus granulosus.<br />

**Thiabendazole is usually inappropriate as first-line therapy for enterobiasis (pinworm). However, when enterobiasis occurs with any of the indications listed for thiabendazole, additional therapy is not<br />

required for most patients.<br />

■ Only indicated when more specific therapy is not available or cannot be used or when further therapy with a second agent is desirable.<br />

4


Table 3. Major Intestinal Parasites, Causative Organisms, and Treatments of Choice 7<br />

Intestinal<br />

Nematodes<br />

Tissue<br />

Nematodes<br />

Cestodes<br />

Trematodes<br />

Major Parasite Infections<br />

Infection (common name) Organism Drug(s) of Choice<br />

Ascariasis 1 (Roundworm) Ascaris lumbricoides Mebendazole, Pyrantel pamoate or Diethylcarbamazine<br />

Uncinariasis (Hookworm) Ancylostoma duodenale Mebendazole or Pyrantel pamoate 2<br />

Necator americanus<br />

Strongyloidiasis (Threadworm)<br />

Strongyloides stercoralis Thiabendazole<br />

Trichuriasis (Whipworm) Trichuris trichiura Mebendazole<br />

Enterobiasis 3 (Pinworm) Enterobius vermicularis Mebendazole, Pyrantel pamoate or Albendazole<br />

Capillariasis<br />

Capillaria philippinensis Mebendazole, Thiabendazole or Albendazole<br />

Trichinosis Trichinella spiralis Steroids for severe symptoms plus Thiabendazole,<br />

Albendazole, Flubendazole 6 or Mebendazole 2<br />

Cutaneous larva migrans<br />

(Creeping eruption)<br />

Ancylostoma braziliense<br />

and others<br />

Thiabendazole, Albendazole or Ivermectin 4<br />

Onchocerciasis (River blindness) Onchocerca volvulus Suramin 5 , Diethylcarbamazine or Ivermectin 4<br />

Dracontiasis (Guinea worm)<br />

Angiostrongyliasis (Rat<br />

lungworm)<br />

Dracunculus medinensis Thiabendazole or Mebendazole<br />

Angiostrongylus<br />

cantonensis<br />

Thiabendazole or Mebendazole<br />

Loiasis Loa loa Diethylcarbamazine<br />

Taeniasis (Beef tapeworm) Taenia saginata Praziquantel 2 or Niclosamide 6<br />

(Pork tapeworm) Taenia solium Praziquantel 2 , Niclosamide 6 or Albendazole<br />

Diphyllobothriasis (Fish Diphyllobothrium latum Praziquantel 2 or Niclosamide 6<br />

tapeworm)<br />

Dog tapeworm Dipylidium caninum Praziquantel 2<br />

Hymenolepiasis (Dwarf<br />

tapeworm)<br />

Hydatid cysts<br />

Schistosomiasis<br />

Hymenolepis nana Praziquantel 2 or Niclosamide 6<br />

Echinococcus<br />

granulosus<br />

Schistosoma mansoni<br />

Schistosoma japonicum<br />

Schistosoma<br />

haematobium<br />

Schistosoma mekongi<br />

Albendazole or Praziquantel<br />

Praziquantel or Oxamniquine<br />

Praziquantel<br />

Praziquantel<br />

Praziquantel<br />

Hermaphroditic Flukes<br />

Fasciolopsiasis(Intestinal fluke) Fasciolopsis buski Praziquantel<br />

Clonorchiasis (Chinese liver<br />

fluke)<br />

Heterophyes heterophyes<br />

Metagonimus yokogawai<br />

Clonorchis sinensis<br />

Praziquantel<br />

Praziquantel<br />

Fascioliasis (Sheep liver fluke) Fasciola hepatica Praziquantel or Bithionol 4<br />

Opisthorchiasis (Liver fluke) Opisthorchis viverrini Praziquantel<br />

Paragonimiasis (Lung fluke) Paragonimus<br />

westermani<br />

1 Thiabendazole is also indicated in Ascariasis.<br />

2 Unlabeled use.<br />

3 Thiabendazole is also indicated in Enterobiasis.<br />

4 Available from the CDC.<br />

5 Available from the CDC, although generally not recommended.<br />

6 Not available in the US.<br />

Praziquantel or Bithionol 4 (alternate)<br />

5


IV.<br />

Pharmacokinetic Parameters<br />

Table 4 lists the pharmacokinetic parameters and mechanisms of action of the anthelmintic<br />

antibiotics.<br />

Table 4. Pharmacokinetic Parameters of the Anthelmintic Agents 5-7<br />

Drug Mechanism of Action Bioavailability Protein<br />

Binding<br />

Albendazole<br />

Ivermectin<br />

Mebendazole<br />

Praziquantel<br />

Pyrantel<br />

Pamoate<br />

Thiabendazole<br />

Has an inhibitory effect on<br />

tubulin polymerization,<br />

which results in the loss of<br />

cytoplasmic microtubules.<br />

Binds selectively to<br />

glutamate-gated chloride<br />

ion channels in<br />

invertebrate nerve and<br />

muscle cells, leading to an<br />

increase in the<br />

permeability of the cell<br />

membrane to chloride ions<br />

with hyperpolarization of<br />

the nerve or muscle cell,<br />

resulting in paralysis and<br />

death of the parasite.<br />

Inhibits the formation of<br />

the worms’ microtubules<br />

and irreversibly blocks<br />

glucose uptake, depleting<br />

endogenous glycogen<br />

stores.<br />

Increases cell membrane<br />

permeability, resulting in a<br />

loss of intracellular<br />

calcium, massive<br />

contractions, and paralysis<br />

of the musculature.<br />

Activity further results in<br />

vacuolization and<br />

disintegration of the<br />

schistosome tegument.<br />

Works as a depolarizing<br />

neuromuscular blocking<br />

agent, resulting in spastic<br />

paralysis of the worm.<br />

Vermicidal or vermifugal<br />

activity.<br />

Poorly absorbed;<br />

systemic activity is<br />

due to the<br />

metabolite<br />

albendazole<br />

sulfoxide. Oral<br />

bioavailability is<br />

higher by up to 5<br />

fold when given<br />

with a fatty meal.<br />

Plasma<br />

concentrations are<br />

proportional to the<br />

dose.<br />

Poorly absorbed<br />

orally<br />

(5-10%)<br />

Rapidly absorbed<br />

(80%)<br />

Metabolis<br />

m<br />

Active<br />

Metabolites<br />

Elimination<br />

70% Liver Yes Biliary;


V. Drug Interactions<br />

There are no level 1 (most severe and life-threatening) drug interactions with the anthelmintic<br />

agents in this class. 8 No drug interactions for ivermectin were documented in any of the resources<br />

used to complete this section of the review, nor where any reported in the manufacturers package<br />

insert.<br />

Table 5. Drug Interactions of the Anthelmintic Agents 8<br />

Drug Significance Interaction Mechanism<br />

Albendazole Level 4 (moderate,<br />

possible)<br />

Albendazole and grapefruit juice Inhibition of albendazole metabolism (CYP3A4) in<br />

the small intestine by grapefruit juice is suspected,<br />

increasing plasma concentrations of albendazole,<br />

and increasing the risk of side-effects.<br />

Mebendazole Level 4 (delayed,<br />

moderate, possible)<br />

Mebendazole and carbamazepine Mechanism is unknown. The pharmacologic<br />

effects of mebendazole may be decreased.<br />

Mebendazole Level 4 (delayed, Mebendazole and hydantoins Mechanism is unknown. The pharmacologic effects<br />

Praziquantel<br />

Praziquantel<br />

Praziquantel<br />

Praziquantel<br />

Thiabendazole<br />

moderate, possible)<br />

Level 2 (delayed,<br />

moderate, probable)<br />

Level 4 (rapid,<br />

moderate, possible)<br />

Level 4 (rapid,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 2 (delayed,<br />

Moderate, suspected)<br />

Praziquantel and cimetidine<br />

Praziquantel and carbamazepine<br />

Praziquantel and hydantoins<br />

Praziquantel and grapefruit juice<br />

Thiabendazole and theophyllines<br />

(xanthine derivatives)<br />

of mebendazole may be decreased.<br />

Cimetidine may inhibit the first-pass metabolism of<br />

praziquantel causing plasma concentrations of<br />

praziquantel to be elevated. This increases both the<br />

effectiveness and risk of adverse reactions.<br />

Mechanism is unknown. Serum praziquantel<br />

concentrations may be reduced, possibly leading to<br />

treatment failures.<br />

Mechanism is unknown. Serum praziquantel<br />

concentrations may be reduced, possibly leading to<br />

treatment failures.<br />

Inhibition of intestinal first-pass metabolism of<br />

praziquantel by grapefruit juice is suspected.<br />

Plasma concentrations of praziquantel may be<br />

elevated, increasing the pharmacologic and adverse<br />

effects.<br />

The exact mechanism is unknown, however,<br />

metabolic inhibition is suspected, and can lead to<br />

increased theophylline serum levels with possible<br />

toxicity. Theophylline levels should be monitored.<br />

Other interactions (per manufacturers labeling): 5<br />

• Pyrantel and piperazine = These drugs have antagonistic modes of action and, therefore, they<br />

should not be administered concomitantly.<br />

• Mebendazole and cimetidine = Preliminary evidence suggests that cimetidine inhibits<br />

mebendazole metabolism and may result in increased plasma concentrations of the drug.<br />

• Albendazole and cimetidine (precipitant drug) = Albendazole sulfoxide concentrations in bile<br />

and cystic fluid were increased ( 2-fold) in hydatid cyst patients treated with cimetidine.<br />

• Albendazole and praziquantel (precipitant drug) = Praziquantel (40 mg/kg) increased mean<br />

maximum plasma concentration and AUC of albendazole sulfoxide by 50% in healthy<br />

subjects.<br />

• Albendazole and dexamethasone (precipitant drug) = Steady-state trough concentrations of<br />

albendazole sulfoxide were 56% higher when 8 mg dexamethasone was coadministered with<br />

each dose of albendazole (15 mg/kg/day) in eight neurocysticercosis patients.<br />

7


VI.<br />

Adverse Drug Events of the Anthelmintic Agents<br />

Albendazole adverse events, although usually mild, differ between hydatid disease and<br />

neurocysticercosis. Treatment discontinuations are predominantly due to leucopenia (0.7%) or<br />

hepatic abnormalities (3.8% in hydatid disease). In addition to the events reported in Table 6,<br />

acute renal failure related to albendazole therapy has been observed, as well as allergic reactions,<br />

rash urticaria, and other hematologic adverse events.<br />

In comparative trials, patients treated with ivermectin experienced more abdominal distention and<br />

chest discomfort than patients treated with albendazole. However, ivermectin was better tolerated<br />

than thiabendazole in comparative studies involving 37 patients treated with thiabendazole. 5<br />

Adverse events may also be more frequent and/or serious in patients with a heavy worm burden.<br />

Historical data have also shown that microfilaricidal drugs, such as diethylcarbamazine citrate,<br />

might cause cutaneous and/or systemic reactions of varying severity, known as the Mazzotti<br />

reaction. This reaction can involve ophthalmological reactions in patients with onchocerciasis. It<br />

is believed the reactions are due to allergic and inflammatory responses to the death of<br />

microfilariae.<br />

Table 6. Common Adverse Events (%) Reported for the Anthelmintic Agents 5-7<br />

Adverse Event<br />

Albendazole<br />

(Hydatid Ds)<br />

Albendazole<br />

(Neurocysticercosis)<br />

Ivermectin Mebendazole Praziquantel Pyrantel Thiabendazole<br />

Body as a Whole<br />

- - - -<br />

- -<br />

Malaise<br />

b<br />

Cardiovascular<br />

- - - - - -<br />

Edema<br />

Hypotension<br />

b<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

6.0<br />

0<br />

0.9<br />

b<br />

b<br />

b<br />

b<br />

Nausea / Vomiting<br />

3.7<br />

6.2<br />

1.8/0.9<br />

b<br />

b<br />

b<br />

Diarrhea<br />

1.8<br />

b<br />

b<br />

b<br />

Epigastric distress<br />

b<br />

Appetite decrease<br />

0.9<br />

b<br />

b<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

1.2<br />


VII.<br />

Dosing and Administration for the Anthelmintic Agents<br />

Table 7. Dosing for the Anthelmintic Agents 5-7<br />

Drug Availability Dose /Frequency/Duration<br />

Albendazole 200mg oral tablet Dosing depends on which parasitic infection is being treated.<br />

Hydatid Disease<br />

60kg or greater: 400mg BID with meals (28-day cycle followed by a 14-day<br />

albendazole-free interval, for a total of 3 cycles).<br />

85 150micrograms/kg<br />

Mebendazole 100mg chewable tablets If a patient is not cured 3 weeks after treatment, a second course of treatment is advised.<br />

The same dose and dose schedule applies to children and adults.<br />

9


Pinworm Whipworm Common Roundworm Hookworm<br />

Dose: 1 tablet, 1 tablet, 1 tablet, 1 tablet,<br />

Once. morning morning morning<br />

and evening and evening and evening<br />

for 3 consecutive for 3 consecutive for 3 consecutive<br />

days. days. days.<br />

Praziquantel 600mg oral tablet Schistosomiasis:<br />

3 doses of 20mg/kg of bodyweight as a 1 day treatment.<br />

Clonorchiasis and Opisthorchiasis:<br />

3 doses of 25mg/kg of bodyweight as a 1 day treatment.<br />

The interval between the doses should not be < 4 and not > 6 hours.<br />

Tablets should be swallowed unchewed with some liquid during meals. Keeping the<br />

tablets or the segments thereof in the mouth may reveal a bitter taste that can produce<br />

gagging or vomiting.<br />

Pyrantel Pamoate<br />

Thiabendazole<br />

Soft gel 180mg<br />

capsules, 50mg/ml oral<br />

suspension, 50mg/ml<br />

liquid<br />

Oral Suspension<br />

500mg/5ml, oral<br />

chewable 500mg tablets<br />

Segments are broken off by pressing the score (notch) with thumbnails. If ¼ of a tablet<br />

is required, this is best achieved by breaking the segment from the outer end.<br />

Maximum total dose is 1 g. Dosage in mg/kg is the same for adults and children.<br />

Nematode infections:<br />

11mg/kg (max. 1gram) given as a single dose; the dose should be repeated after 2 weeks<br />

in patients with enterobiasis.<br />

Intestinal Hookworm (Ancylostoma duodenale or Necator americanus) or<br />

eosinophilic enterocolitis (Ancylostoma caninum):<br />

11mg / kg (max. 1 gram) once daily for 3 consecutive days. A repeated stool<br />

examination should be performed 2 weeks after treatment and the regimen should be<br />

repeated if results are positive.<br />

Moniliformis:<br />

11mg / kg given once; this dosage should be repeated twice at 2-week intervals.<br />

May be administered without regard to ingestion of food or time of day. Purging is not<br />

necessary. May be taken with milk or fruit juices.<br />

The usual dosage schedule for all conditions is 2 doses per day. Maximum daily dose is<br />

3 g after meals if possible. Dose is determined by body weight.<br />

Weight Each Dose (g) ml<br />

30lb 0.25g (1/2 tablet) 2.5ml (1/2 teaspoon)<br />

50lb 0.5g (1 tablet) 5.0ml (1 teaspoon)<br />

75lb 0.75g (1.5 tablets) 7.5ml (1.5 teaspoons)<br />

100lb 1.0g (2 tablets) 10ml (2 teaspoons)<br />

125lb 1.25g (2.5 tablets) 12.5ml (2.5 teaspoons)<br />

150lb & over 1.5g (3 tablets) 15ml (3 teaspoons)<br />

Regimens Based on Indication<br />

Indication Regimen Comments<br />

Strongyloidiasis 1 2 doses/day for 2 successive days. May also use<br />

Ascariasis 1<br />

single dose of<br />

Uncinariasis 1<br />

20mg/lb (44mg/kg)<br />

Trichuriasis 1<br />

but with higher sideeffects.<br />

Cutaneous larva migrans 2 doses/day for 2 successive days. A second course is<br />

(creeping eruption)<br />

recommended if<br />

if active lesions are still<br />

10


present 2 days after end<br />

of therapy.<br />

Trichinosis 1 2 doses/day for 2-4 successive days. Optimal dosage has not<br />

Individualize dosage.<br />

been established.<br />

Visceral larva migrans 2 doses/day for 7 successive days. Safety and efficacy data<br />

On the 7 day treatment<br />

are limited.<br />

1<br />

Clinical experience with thiabendazole in children weighing < 13.6 kg (30 lbs) is limited.<br />

Special Dosing Considerations<br />

Table 8. Special Dosing Considerations for the Anthelmintic Agents 5-7, 9<br />

Drug Renal<br />

Dosing<br />

Hepatic<br />

Dosing<br />

Pediatric Use Pregnancy<br />

Category<br />

Albendazole No No Experience in age


VIII. Comparative Effectiveness of the Anthelmintic Agents<br />

Not all of the agents in this class can be used for the same helminthic infections.<br />

Table 9. Additional Outcomes Evidence for the Anthelmintics<br />

Study Sample Treatment /<br />

Duration<br />

Albanese<br />

G, et al. 12 n=56 Albendazole,<br />

thiabendazole and<br />

combinations with<br />

cryotherapy<br />

Muchiri<br />

EM, et<br />

al. 13 n=1,186 4 and 6 month<br />

repeated treatment<br />

with albendazole or<br />

mebendazole<br />

Legesse<br />

M, et al. 14 - Mebendazole 100mg<br />

BID for 3 days vs.<br />

albendazole 400mg<br />

single dose<br />

Legesse Four<br />

M, et al. 15 treatment<br />

Albendazole 400mg<br />

single dose vs. three<br />

Results<br />

In comparing albendazole, thiabendazole, and combinations of both drugs with<br />

cryotherapy for the treatment of creeping eruption caused by nematodes of the<br />

Ancylostoma species:<br />

• Thirteen patients received cryotherapy alone, one received<br />

thiabendazole plus cryotherapy, six received thiabendazole, two<br />

received albendazole plus cryotherapy, and 34 received albendazole<br />

alone.<br />

• A prompt and definitive cure was achieved in all 56 patients.<br />

• The therapeutic effectiveness of the various methods used is<br />

equivalent.<br />

• The authors in this study felt albendazole should be the first choice for<br />

treatment due to the drug’s tolerability and favorable patient<br />

compliance.<br />

In evaluating the effectiveness of four and six month repeated treatment with<br />

albendazole 600mg or mebendazole 600mg on helminth infections on children<br />

(age 4-19) in six primary schools in Kenya:<br />

• Overall, albendazole produced better cure rates and egg reduction rates<br />

for geohelminths. The cure rates for albendazole were 92.4% for<br />

hookworm infection, 83.5% for Ascaris lumbricoides, and 67.8% for<br />

Trichuris trichiura.<br />

• Mebendazole given either 2 or 3 times in a year had cure rates of 50<br />

and 55.0% (respectively) for hookworm, 79.6 and 97.5% for A.<br />

lumbricoides, and 60.6 and 68.3% for T. trichiura infection.<br />

• The geometric mean intensity of hookworm eggs per gram (epg) of<br />

stool decreased by 96.7% after albendazole treatment compared with<br />

66.3 and 85.1%, respectively, for 2 or 3 doses of mebendazole (p <<br />

0.05) over the same period.<br />

• Reductions in epg for A. lumbricoides and T. trichiura were<br />

comparable for both drugs. Results indicate that treatment with<br />

albendazole at a six month interval was more effective than<br />

mebendazole regimens and may be the best choice for use in the<br />

control of the three geohelminths.<br />

In comparing mebendazole versus albendazole for the treatment of single or<br />

mixed Ascaris lumbricoides and Trichuris trichiura:<br />

• Both drugs were found to be highly effective against Ascaris<br />

lumbricoides infection, with cure rate of over 96% and egg reduction<br />

of over 99.8%.<br />

• The efficacy of the two drugs against Trichuris trichiura infection was<br />

low.<br />

• Mebendazole appeared to be more effective against Trichuris trichiura<br />

in that it exhibited a cure rate of 34.7% and egg reduction of 92.3% as<br />

opposed to albendazole, which exhibited a cure rate and egg reduction<br />

rate of 13.9% and 63.4%, respectively.<br />

• The two drugs appeared to have little effect on Schistosoma mansoni<br />

infection.<br />

• More complaints were reported by individuals treated with<br />

albendazole than with mebendazole.<br />

• Summary: mebendazole appears to be safer and more effective for the<br />

treatment of single or mixed infections with Trichuris trichiura and<br />

Ascaris lumbricoides as compared to albendazole.<br />

In evaluating albendazole and three brands of mebendazole for the treatment of<br />

single or mixed Ascaris lumbricoides and Trichuris trichiura:<br />

12


groups<br />

of school<br />

children<br />

brands of<br />

mebendazole at doses<br />

of 100mg BID for 3<br />

days<br />

Datry A,<br />

et al. 16 n=60 Albendazole<br />

400mg/day for 3 days<br />

vs. ivermectin 150-<br />

200 micrograms/kg in<br />

a single dose<br />

Tabi TE, n=99 Albendazole 400mg<br />

et al. 17 for 3 days vs. placebo<br />

Simonsen<br />

PR, et<br />

al. 18 n=1,829 Ivermectin 150-200<br />

micrograms/kg alone<br />

or in combination<br />

with albendazole<br />

400mg<br />

• The percentage cure rate and egg reduction rate obtained with<br />

albendazole and mebendazole from the three brands were not<br />

significantly different in the treatment of ascariasis.<br />

• However, significant differences were found among the percentage<br />

cure rates and egg reduction rates of the four treatment groups in the<br />

treatment of trichuriasis. Comparatively, high cure rate (89.8%) and<br />

egg reduction rate (99.1%) were observed in the Vermox (Janssen)<br />

treated group followed by the Unibios (India) treated group (53.3%<br />

and 96.53% cure and egg reduction rates, respectively), whereas low<br />

cure rate (17.1%) and egg reduction rate (69.8%) were seen in the<br />

albendazole treated group.<br />

• The results of this study suggest that in areas where single or mixed<br />

infections with Trichuris trichiura and/or Ascaris lumbricoides are<br />

common, this disease is a public health problem. When laboratory<br />

facilities are not available to make parasite identification, mebendazole<br />

would be the drug of choice to treat trichuriasis and ascariasis.<br />

• However, either mebendazole from the different brands or albendazole<br />

is effective in the treatment of ascariasis in areas where trichuriasis is<br />

not prevalent.<br />

In a randomized trial comparing ivermectin with albendazole in patients with<br />

confirmed Strongyloides stercoralis infection:<br />

• Parasitological cure was obtained in 24 of the 29 patients treated with<br />

ivermectin (83%) and in 9 of the 24 patients who were given<br />

albendazole (38%); ivermectin was significantly more effective than<br />

albendazole (p < 0.01).<br />

• Clinical and biological adverse reactions were negligible in both<br />

treatment groups.<br />

• The 20 patients who failed therapy were given a second treatment<br />

course with 150-200 micrograms/kg of ivermectin in a single dose or on<br />

two consecutive days. Sixteen patients were cured and the other four<br />

had only incomplete follow-up.<br />

• Ivermectin therefore constitutes an acceptable therapeutic alternative for<br />

uncomplicated strongyloidiasis.<br />

In this double-blind, placebo-controlled, crossover clinical trial of albendazole<br />

vs. placebo for the treatment of loiasis:<br />

• Because of life-threatening, post-treatment reactions in patients with<br />

loiasis treated with ivermectin, other treatments are necessary.<br />

• Patients received treatment for three days and were followed for 180<br />

days, and then were crossed over and followed for an additional 180<br />

days.<br />

• In those initially receiving albendazole (ALB/PLAC), microfilarial<br />

levels decreased significantly by day 90 (p < 0.043), but returned to<br />

baseline by day 180. In those receiving albendazole at day 180<br />

(PLAC/ALB), microfilarial levels also decreased following<br />

albendazole (p = 0.005).<br />

• Blood eosinophil and antifilarial IgG levels did not change<br />

significantly for either group, although antifilarial IgG4 levels did in<br />

the ALB/PLAC group at day 180.<br />

• Most subjects continued to have elevations in microfilaremia,<br />

suggesting that more intensive regimens of albendazole will be<br />

necessary to reduce Loa microfilaremia to levels safe enough to allow<br />

for ivermectin use.<br />

In evaluating the efficacy of a single dose of ivermectin alone or in combination<br />

with albendazole in children in six primary schools in coastal Tanzania:<br />

• Both treatment regimens resulted in a considerable decrease in mean<br />

microfilaria (mf) intensities, with overall reductions being slightly but<br />

statistically significantly higher for the combination than for<br />

ivermectin alone.<br />

• The difference in effect between the two regimens was most<br />

pronounced at six months, whereas it was minor at 12 months after<br />

13


Makunde - Albendazole 400mg in<br />

al. 19 ivermectin 150mcg/kg<br />

WH, et<br />

combination with<br />

Awadzi<br />

K, et al. 20 n=42 Ivermectin<br />

(200mcg/kg) plus<br />

albendazole (400mg)<br />

vs. ivermectin alone<br />

(200mcg.kg)<br />

Safdar A, n=25 Frequency of<br />

et al. 21 Strongyloides<br />

stercoralis infestation<br />

treatment.<br />

• The relative effect of treatment on mean circulating filarial antigen<br />

(CFA) units was less pronounced than on microfilaria.<br />

• For both treatment regimens, reductions in CFA intensity appeared to<br />

be higher in children who were both CFA and mf positive before<br />

treatment, which may suggest that treatment mainly affected the<br />

survival and/or production of mf, rather than the survival of adult<br />

worms.<br />

• Adverse reactions were few and mild in both groups, and mainly<br />

reported from pre-treatment mf and CFA positive children.<br />

In order to use ivermectin with albendazole for the elimination of lymphatic<br />

filariasis, the potential risk of adverse events in individuals infected with both<br />

lymphatic filariasis and onchocerciasis was compared in this crossover, doubleblind<br />

study (for patients with co-infections):<br />

• Patients with a single infection of bancroftian filariasis underwent an<br />

open design comparing two treatments, where one group received a<br />

single dose of albendazole (400mg) plus ivermectin (150microg/kg)<br />

(Group C) while the other group received a single dose of albendazole<br />

(400mg) alone (Group D).<br />

• For co-infection, one group was allocated a single dose of ivermectin<br />

(150micrograms/kg) plus albendazole (400mg) (Group A). The other<br />

group received placebo (Group B). Five days later the treatment<br />

regimen was reversed, with the Group A receiving placebo and Group<br />

B receiving treatment.<br />

• In individuals co-infected with bancroftian filariasis and<br />

onchocerciasis, treatment with ivermectin and albendazole was safe<br />

and tolerable.<br />

• Physiological indices showed no differences between groups with coinfection<br />

(W. bancrofti and O. volvulus) or single infection (W.<br />

bancrofti).<br />

• The frequency of adverse events in co-infected individuals was 63%<br />

(5/8, Group A, albendazole + ivermectin) and 57% (4/7, Group B,<br />

placebo) with mild or moderate intensity.<br />

• In single W. bancrofti infection the frequency of adverse events was<br />

50% (6/12, Group C, albendazole + ivermectin) and 38% (5/13, Group<br />

D, albendazole) and of a similar intensity to those experienced with coinfection.<br />

• There were no differences in adverse events between treatment groups.<br />

• There was no significant difference in the reduction of microfilaraemia<br />

following treatment with albendazole and ivermectin in groups with<br />

single or co-infection.<br />

This randomized, double-blind, placebo-controlled trial was conducted to<br />

determine if co-administration of ivermectin with albendazole is safe and<br />

effective against Onchocerca volvulus compared to ivermectin alone, and<br />

whether a significant pharmacokinetic interaction occurs:<br />

• The co-administration of ivermectin with albendazole did not produce<br />

more severe adverse effects than ivermectin alone.<br />

• Both nodule examiners found that the combination was not<br />

macrofilaricidal and that it was not clearly superior to ivermectin alone<br />

in the effects on reproductive activity; this was supported by the<br />

similar efficacy of the two regimens in the suppression of skin<br />

microfilariae.<br />

• There was no significant pharmacokinetic interaction.<br />

• Although the co-administration of ivermectin with albendazole appears<br />

safe, it offers no advantage over ivermectin alone in the control of<br />

onchocerciasis.<br />

• The combination does not require an alteration in the dosage of either<br />

component.<br />

This retrospective analysis of S. stercoralis infection in patients with cancer<br />

undergoing cancer treatment showed:<br />

• The overall S. stercoralis infection frequency was approximately 1.0 per<br />

14


and complications in<br />

patients with cancer<br />

between 1971 and<br />

2003<br />

Belizario<br />

VY, et<br />

al. 22 n=784 Single doses of<br />

albendazole,<br />

ivermectin, and<br />

diethylcarbamazine<br />

(all given with<br />

placebo), and of the<br />

combination of<br />

albendazole +<br />

ivermectin and<br />

albendazole +<br />

diethylcarbamazine<br />

10,000 new cancer cases between 1971 and 2003.<br />

• Twenty-two of 25 patients (88%) were U.S. residents (19 from Texas; one<br />

each from Mississippi, Tennessee, and Puerto Rico), and the remaining<br />

three (13%) were from Latin America.<br />

• Thirteen (52%) had solid-organ malignancies, whereas 12 (48%) had<br />

hematologic malignancies (lymphoma or multiple myeloma, n=8; leukemia,<br />

n=3; aplastic anemia, n=1).<br />

• Twelve patients (48%) received systemic corticosteroids, 9 (36%) received<br />

antineoplastic therapy, and 2 underwent hematopoietic stem cell<br />

transplantation (HSCT).<br />

• Diarrhea was reported in 13 patients (57%), and eosinophilia was observed<br />

in 11 patients (48%); four patients (16%) had probable hyperinfection<br />

syndrome (in three cases of polymicrobial gram-negative bacteremia, one<br />

patient had Klebsiella pneumoniae pneumonia, whereas one patient<br />

presented with K. pneumoniae lung infection alone). Evidence of definite<br />

pulmonary hyperinfection syndrome was observed in 2 HSCT recipients<br />

(8%).<br />

• Fourteen (74%) of 19 patients responded to thiabendazole therapy. Two<br />

patients with definite pulmonary hyperinfection syndrome developed fatal<br />

S. stercoralis hemorrhagic alveolitis despite receiving high-dose<br />

thiabendazole plus ivermectin therapy.<br />

• In the current study, strongyloidiasis was uncommon in patients with cancer<br />

and remained localized in individuals with solid-organ malignancies.<br />

Definite pulmonary accelerated autoinfections were observed only in HSCT<br />

recipients.<br />

In evaluating the efficacy of various single dose treatments against the common<br />

intestinal helminthiases caused by Ascaris and Trichuris species in this randomized,<br />

placebo-controlled trial:<br />

• Albendazole, ivermectin and the drug combinations gave significantly<br />

higher cure and egg reduction rates for ascariasis than diethylcarbamazine.<br />

• For trichuriasis, albendazole + ivermectin gave significantly higher cure and<br />

egg reduction rates than the other treatments: the infection rates were lower<br />

180 and 360 days after treatment.<br />

• Summary: Because of the superiority of albendazole + ivermectin against<br />

both lymphatic filariasis and trichuriasis, this combination appears to be a<br />

suitable tool for the integrated or combined control of both public health<br />

programs.<br />

Additional Evidence<br />

Dose Simplification: In most cases, the anthelmintic agents in this class are given as a single dose or for a<br />

brief duration (acute use), depending on the infection being treated. When given for longer durations, dosing<br />

is QD-BID. Therefore, research into dose simplification within this class is not an applicable.<br />

Stable Therapy: Limited clinical data is available on resistance to the anthelmintic medications in humans.<br />

Interestingly, more studies have been published on resistance to helminthic infections in horses, pigs, and<br />

sheep. One study from the region of Mali showed that in evaluating a standard egg hatch assay, patients from<br />

Mali with N. americanus (hookworm) infections were more resistant to benzimidazoles compared with a<br />

laboratory-maintained strain that had not been exposed to anthelmintics, suggesting increased drug resistance<br />

from drug failures. In this study, single-dose mebendazole was ineffective against hookworm infections in<br />

Mali. 23 Although the clinical evidence does not suggest problems with resistance patterns in the U.S,<br />

resistance to the anthelmintic drugs should be considered in patients who have previously been treated for<br />

helminthic infections and in those with apparent treatment failure.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal clinical literature<br />

relevant to use of the anthelmintics and their impact on physician visits.<br />

15


IX.<br />

Conclusions<br />

Mebendazole and pyrantel pamoate are available to patients due to their over-the-counter status and generic<br />

availability. Mebendazole and pyrantel can be used to treat pinworm, whipworm, roundworm, and<br />

hookworm infections (pyrantel = pinworm and roundworm only), which are the more common helminthic<br />

infections found in the United States. Mebendazole and pyrantel are the treatments of choice for these<br />

helminthic infections.<br />

Other less common helminthic infections in the United States include strongyloides, schistosoma, liver<br />

flukes, onchocerciasis, neurocysticercosis, and hydatid disease. These infections are rarely found in the U.S,<br />

but may occur in immunocompromised patients or in patients who have traveled to endemic areas. At this<br />

time, treatment of these helminthic infections is considered outside the scope of general use of the drugs<br />

within this class. Because albendazole, ivermectin, praziquantel and thiabendazole have indications for these<br />

infections, they should be available for special needs/circumstances that require medical justification through<br />

the prior authorization process. Proper medical justification will provide patient access to these agents.<br />

For these reasons, mebendazole and pyrantel pamoate offer clinical advantages when used for their respective<br />

treatment indications. These agents are available OTC and with generic formulations. The remaining<br />

anthelmintic drugs in this class (albendazole, ivermectin, praziquantel and thiabendazole) are comparable to<br />

each other and to the generics and OTC products in this class and offer no significant clinical advantage over<br />

other alternatives in general use.<br />

X. Recommendations<br />

No brand anthelmintic is recommended for preferred status.<br />

16


References<br />

1. Anandan JV. Parasitic Diseases. In: <strong>Pharmacotherapy</strong>. A Pathophysiologic Approach, Fifth Edition.<br />

Dipiro JT, Talbert RL, Yee GC, et al. Eds. McGraw-Hill. New York. 2002. Pg. 1967-581980.<br />

2. Hall A. Anthelmintics: drugs for treating worms. Afr Health 1998 Sep;20(6):4-6.<br />

3. Centers for Disease Control and Prevention (CDC). Guidelines for preventing opportunistic infections<br />

among hematopoietic stem cell transplant recipients. MMWR Oct 20 2000;49;RR-10.<br />

4. MMWR. Palmar pallor as an indicator for anthelmintic treatment among ill children aged 2-4 years-western<br />

Kenya, 1998. JAMA June 14 2000;283(22):2925-2927.<br />

5. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58, 2004.<br />

Thomson PDR. Montvale, NJ. 2004.<br />

6. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American Society of<br />

Health-System Pharmacists. Bethesda. 2004.<br />

7. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

8. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

9. Briggs GG, Freeman RK, Yaffe SJ. Drugs in pregnancy and lactation. A reference guide to fetal and<br />

neonatal risk. Sixth Edition. Lippincott, Williams, & Wilkins, Philadelphia, 2002.<br />

10. Tarr PE, Miele PS, Peregory KS, et al. Case report: rectal administration of ivermectin to a patient with<br />

Strongyloides hyperinfection syndrome. Am J Trop Med Hyg Apr 2003;68(4):453-455.<br />

11. Chiodini PL, Reid AJC, Wiselka MJ, et al. Parenteral ivermectin in strongyloides hyperinfection. Lancet Jan<br />

1, 2000;355(9197):43-44.<br />

12. Albanese G, Venturi C, Galbiati G, et al. Treatment of larva migrans cutanea (creeping eruption): a<br />

comparison between albendazole and traditional therapy. Int J Dermatol 2001 Jan;40():67-71.<br />

13. Muchiri EM, Thiong’o FW, Mangnussen P, et al. A comparative study of different albendazole and<br />

mebendazole regimens for the treatment of intestinal infections in school children of Usigu Division, western<br />

Kenya. J Parasitol 2001 Apr;87(2):413-8.<br />

14. Legesse M, Erko B, Medhin G. Efficacy of albendazole and mebendazole in the treatment of Ascaris and<br />

Trichuris infections. Ethiop Med J 2002 Oct;40(4):335-43.<br />

15. Legesse M, Erko B, Medhin G. Comparative efficacy of albendazole and three brands of mebendazole in the<br />

treatment of ascariasis and trichuriasis. East Afr Med J 2004 Mar;81(3):134-8.<br />

16. Datry A, Hilmarsdottir I, Mayorga-Sagastume R, et al. Treatment of Strongyloides stercoralis infection with<br />

ivermectin compared with albendazole: results of an open study of 60 cases. Trans R Soc Trop Med Hyg<br />

1994 May-Jun:88(3):344-5.<br />

17. Tabi TE, Befidi-Mengue R, Nutman TB, et al. Human loiasis in a Camemroonian village: a double-blind,<br />

placebo-controlled, crossover clinical trial of a three-day albendazole regimen. Am J Trop Med Hyg 2004<br />

Aug;71(2):211-215.<br />

18. Simonsen PE, Magesa SM, Dunyo SK, et al. The effect of single dose ivermectin alone or in combination<br />

with albendazole on Wuchereria bancrofti infection in primary school children in Tanzania. Trans R Soc<br />

Trop med Hyg 2004 Aug;98(8):462-72.<br />

19. Makunde WH, Kamugisha LM, Massaga JJ, et al. Treatment of co-infection with bancroftian filariasis and<br />

onchocerciasis: a safety and efficacy y study of albendazole with ivermectin compared to treatment of single<br />

infection with bancroftian filariasis. Filaria J 2003 Nov 6;2(1):15.<br />

20. Awadzi K, Edwards G, Duke BO, et al. The co-administration of ivermectin and albendazole—safety,<br />

pharmacokinetics and efficacy against Onchocerca volvulus. Ann Trop Med Parasitol 2003 Mar;97(2):165-<br />

78.<br />

21. Safdar A, Malathum K, Rodriguez SJ, et al. Strongyloidiasis in patients at a comprehensive cancer center in<br />

the United States. Cancer 2004 Apr 1;100(7):1531-6.<br />

22. Belizario VY, Amarillo ME, de Leon WU, et al. A comparison of the efficacy of single doses of albendazole,<br />

ivermectin, and diethylcarbamazine alone or in combinations against Ascaris and Trichuris species. Bull<br />

World Health Organ 2003;81(1):35-42.<br />

23. De Clercq D, Sacko M, Behnke J, et al. Failure of mebendazole in treatment of human hookworm infections<br />

in the southern region of Mali. Am J Trop Hyg 1997 Jul;57(1):25-30.<br />

17


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Aminoglycosides<br />

AHFS 081202<br />

October 27, 2004<br />

I. Overview<br />

The aminoglycosides are generally active against aerobic gram-negative bacteria and gram-positive bacteria<br />

and are used for serious infections, including septicemia (including neonatal sepsis), bone and joint<br />

infections, skin and soft-tissue infections, respiratory tract infections, uncomplicated urinary tract infections,<br />

and postoperative and intra-abdominal infections. The drugs in this class work by creating fissures in the<br />

outer membrane of the bacterial cell. 1 Due to in vitro synergism, concomitant use of an aminoglycoside with<br />

an extended-spectrum penicillin with antipseudomonal activity, has been recommended. 2 However, in vitro<br />

inactivation of aminoglycosides by β-lactam antibiotics indicates that the drugs should be administered<br />

separately and in vitro mixing of these drugs should be avoided.<br />

Aminoglycosides are also used in conjunction with a β-lactam antibiotic, a carbapenem, an extendedspectrum<br />

penicillin, or a fixed combination of an extended-spectrum penicillin and a β-lactamase inhibitor for<br />

empiric anti-infective therapy of presumed bacterial infections in febrile neutropenic patients. Additionally,<br />

most strains of enterococci are resistant to aminoglycosides alone, and penicillin therapy alone is usually<br />

inadequate in infections caused by these organisms; however, due to synergism, gentamicin or streptomycin<br />

used together with penicillin G or ampicillin is often effective in the treatment of enterococcal endocarditis. 2<br />

Gentamicin and ampicillin can be used for prophylaxis of bacterial endocarditis in adults and children<br />

undergoing certain genitourinary or nonesophageal GI tract procedures likely to cause transient bacteremia.<br />

The American Academy of Pediatrics states that in invasive enterococcal infections, including endocarditis<br />

and meningitis, ampicillin and vancomycin combined with an aminoglycoside (usually gentamicin) should be<br />

administered until in vitro susceptibility is known and appropriate combination therapy can be selected. 2 The<br />

Centers for Disease Control and Prevention suggest a regimen of IM or IV gentamicin in combination with<br />

IV clindamycin as one possible parenteral regimen for the treatment of acute pelvic inflammatory disease<br />

(PID).<br />

In general, the choice of a specific parenteral aminoglycoside should be based on the usual spectrum and<br />

pattern of aminoglycoside resistance in the hospital or community until results of in vitro tests are available.<br />

Generally, when given a susceptible organism, amikacin, gentamicin, kanamycin, and tobramycin appear to<br />

be equally effective when administered in appropriate doses. Resistance to the aminoglycosides is rare, but<br />

increasing in frequency. 1 Amikacin may be effective in infections caused by gentamicin, kanamycin, and/or<br />

tobramycin resistant organisms, especially P. rettgeri, P. stuartii, S. marcescens, K. pneumoniae, and Ps.<br />

Aeruginosa. However, there are also strains of bacteria resistant to amikacin that may be susceptible to<br />

gentamicin and/or tobramycin.<br />

In looking at oral use of the aminoglycosides, kanamycin and neomycin are both used orally for preoperative<br />

intestinal antisepsis as an adjunct to mechanical cleansing of the large intestines. Regimens of oral neomycin<br />

and oral erythromycin are sometimes used for perioperative prophylaxis in patients undergoing colorectal<br />

surgery. Neomycin is also used orally as an adjunct to fluid and electrolyte replacement in the treatment of<br />

severe diarrhea caused by susceptible strains of enteropathogenic E. coli.<br />

Finally, the aminoglycosides can be used when administered by oral inhalation for the management of<br />

bronchopulmonary Ps. aeruginosa infections in cystic fibrosis patients. This type of treatment is generally<br />

long-term suppressive therapy for prophylaxis of exacerbations of infections and is not routinely<br />

recommended for the treatment of acute exacerbations.<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drugs in this review.<br />

18


Table 1. Aminoglycosides in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name<br />

Amikacin Sulfate Parenteral Amkin*, Amkin Pediatric*<br />

Gentamicin Sulfate Parenteral Garamycin*<br />

Kanamycin Sulfate Parenteral, Oral Kantrex* (oral is brand only)<br />

Neomycin Sulfate Oral Neo-Fradin* (oral solution is brand only)<br />

Streptomycin Sulfate Parenteral Streptomycin*<br />

Tobramycin Sulfate<br />

*Generic Available.<br />

Oral Inhalation,<br />

Parenteral<br />

Nebcin* (injection is generic, powder for<br />

injection is brand), Tobi<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Despite the introduction of newer, less toxic antimicrobial agents, aminoglycosides continue to serve a useful<br />

role in the treatment of serious enterococcal and gram-negative bacillary infections. 1 According to an article<br />

from the American Academy of Family Physicians, gentamicin is the aminoglycoside used most often<br />

because of its reliable activity against gram-negative aerobes, and in general, gentamicin, tobramycin, and<br />

amikacin are used in similar circumstances, often interchangeably. 1, 3 Tobramycin may be the<br />

aminoglycoside of choice for use against P. aeruginosa because it has shown greater in vitro activity.<br />

Amikacin is particularly effective when used against bacteria that are resistant to other aminoglycosides,<br />

since its chemical structure makes it less susceptible to inactivating enzymes. 3 Depending on local resistance<br />

patterns, amikacin may be the preferred agent for serious nosocomial infections caused by gram-negative<br />

bacilli.<br />

When selecting anti-infectives for prophylaxis of recurrent rheumatic fever or prophylaxis of bacterial<br />

endocarditis, the current recommendations published by the American Heart Association should be consulted.<br />

Additionally, hospital and community resistance patterns are important considerations. The following table<br />

highlights drugs of choice for multiple types of microorganisms.<br />

Table 2. Aminoglycosides as Drugs of Choice 4<br />

Microorganism<br />

Drug<br />

Gram-Positive Cocci Enterococcus faecalis, serious infections (endocarditis,<br />

meningitis, pyelonephritis with bacteremia)<br />

Ampicillin (or penicillin G) +<br />

gentamicin or streptomycin<br />

Streptococcus, viridans group<br />

Penicillin G ± gentamicin a<br />

Staphylococcus aureus / Staphylococcus epidermidis, Vancomycin ± gentamicin or rifampin<br />

methicillin (oxacillin) –resistant<br />

Gram-Negative Cocci - -<br />

Gram-Positive Bacilli - -<br />

Gram-Negative Bacilli Acinetobacter species Imipenem or meropenem either ±<br />

aminoglycoside (per sensitivities;<br />

amikacin usually most effective)<br />

Enterobacter species<br />

Imipenem, meropenem, or cefepime<br />

plus aminoglycoside (per sensitivities)<br />

Klebsiella pneumoniae (UTI only)<br />

Aminoglycoside (per sensitivities)<br />

Pseudomonas aeruginosa<br />

UTI only<br />

Piperacillin or ceftazidime plus<br />

aminoglycoside (per sensitivities)<br />

Aminoglycoside (per sensitivities)<br />

Serratia marcescens Third-generation cephalosporin ±<br />

gentamicin<br />

Misc. Microorganisms - -<br />

Spirochetes - -<br />

a<br />

Gentamicin should be added if tolerance or moderately susceptible (MIC >0.1g/ml) organisms are encountered; streptomycin is used but may be more<br />

toxic.<br />

19


Additionally, the following table reflects susceptibilities to the aminoglycosides.<br />

Table 3. Organisms Generally Susceptible to Aminoglycosides 5, 6<br />

Organisms Amikacin Gentamicin Kanamycin Streptomycin Tobramycin<br />

Gram-positive Mycobacterium tuberculosis 1 2<br />

Staphylococci 3 3<br />

S. aureus 3<br />

S. epidermidis<br />

Streptococci 2<br />

S. faecalis 2 2 2<br />

Gram-negative Acinetobacter sp.<br />

Brucella sp.<br />

Citrobacter sp.<br />

Enterobacter sp.<br />

Escherichia coli<br />

Hemophilus influenzae 2<br />

Hemophilus ducreyi<br />

Klebsiella sp. 2<br />

Morganella morganii<br />

Neisseria sp.<br />

Proteus sp. 4 4 4 4<br />

Providencia sp.<br />

Pseudomonas sp.<br />

P. aeruginosa 2<br />

Salmonella sp.<br />

Serratia sp.<br />

Shigella sp.<br />

Yersinia (Pasteurella) pestis<br />

1<br />

= generally susceptible<br />

3 Penicillinase-producing and nonpenicillinase-producing.<br />

2 Usually used concomitantly with other anti-infectives.<br />

4 Indole-positive and indole-negative.<br />

20


III.<br />

Comparative Indications of the Aminoglycoside Antibiotics<br />

Aminoglycosides should be reserved for treatment of infections caused by organisms not sensitive to less toxic agents. Safety for treatment<br />

periods greater than 14 days has not been established and many of the aminoglycosides should not be used long-term due to adverse events.<br />

Table 4. FDA-Approved Indications for the Aminoglycosides 2, 6<br />

Drug Infections (General) Gram Negative<br />

Infections<br />

Amikacin Sulfate<br />

✔<br />

Bacterial septicemia; infections of<br />

the respiratory tract, joints, bones,<br />

skin and soft tissue; meningitis;<br />

intra-abdominal infections; burns;<br />

post-operative infections;<br />

complicated and recurrent UTI’s.<br />

✔<br />

Consider as initial therapy in<br />

suspected gram-negative<br />

infections; therapy may be<br />

instituted before obtaining<br />

the results of susceptibility<br />

testing. It is effective in<br />

infections caused by<br />

gentamicin- or tobramycinresistant<br />

strains of gramnegative<br />

organisms,<br />

particularly P. rettgeri, P.<br />

stuartii, S. marcescens and<br />

P. aeruginosa.<br />

Staphylococcal Infections Neonatal Sepsis Intrathecal<br />

Administration<br />

✔<br />

Initial therapy under certain<br />

conditions in the treatment of<br />

known or suspected<br />

staphylococcal disease, such as:<br />

Severe infections where the<br />

causative organism may be either<br />

a gram-negative bacterium or a<br />

staphylococcus; infections<br />

caused by susceptible strains of<br />

staphylococci in patients allergic<br />

to other antibiotics; and in mixed<br />

staphylococcal/gram-negative<br />

infections.<br />

✔<br />

In severe infections,<br />

concomitant therapy<br />

with a penicillin-type<br />

drug may be indicated<br />

because of the<br />

possibility of<br />

infections caused by<br />

gram-positive<br />

organisms, such as<br />

streptococci or<br />

pneumococci.<br />

Other<br />

- -<br />

Gentamicin Sulfate<br />

✔<br />

Serious infections caused by<br />

susceptible strains of<br />

Pseudomonas aeruginosa,<br />

Proteus sp. (indole-positive and<br />

indole-negative), Escherichia coli,<br />

Klebsiella sp., Enterobacter sp.,<br />

Serratia sp., Citrobacter sp. and<br />

Staphylococcus sp. (coagulasepositive<br />

and coagulase-negative).<br />

Effective in bacterial neonatal<br />

sepsis; bacterial septicemia;<br />

meningitis, urinary tract,<br />

respiratory tract, GI tract, skin,<br />

bone and soft tissue (including<br />

burns).<br />

✔<br />

Consider as initial therapy in<br />

suspected or confirmed<br />

gram-negative infections.<br />

✔<br />

While not the antibiotic of first<br />

choice, consider gentamicin<br />

when penicillins or other less<br />

toxic drugs are contraindicated,<br />

when bacterial susceptibility<br />

tests and clinical judgment<br />

indicate its use and in mixed<br />

infections caused by susceptible<br />

strains of staphylococci and<br />

gram-negative organisms.<br />

✔<br />

✔<br />

Indicated as<br />

adjunctive therapy to<br />

systemic gentamicin<br />

in the treatment of<br />

serious CNS<br />

infections<br />

(meningitis,<br />

ventriculitis) caused<br />

by susceptible<br />

Pseudomonas sp.<br />

-<br />

21


Drug Infections (General) Gram Negative<br />

Infections<br />

Kanamycin Sulfate<br />

✔<br />

✔<br />

Initial therapy for one or more of Indicated for initial therapy<br />

the following: Escherichia coli, of severe infections thought<br />

Proteus sp. (both indole-positive to be susceptible in patients<br />

and indole-negative), allergic to other antibiotics,<br />

Enterobacter aerogenes,<br />

or in mixed<br />

Klebsiella pneumoniae, Serratia staphylococcal/gramnegative<br />

marcescens and Acinetobacter sp.<br />

infections.<br />

Also initial therapy with a<br />

penicillin or cephalosporin before<br />

obtaining results of susceptibility<br />

testing.<br />

Staphylococcal Infections Neonatal Sepsis Intrathecal<br />

Administration<br />

Not the drug of choice for<br />

staphylococcal infections.<br />

✔ -<br />

Other<br />

Oral only:<br />

Suppression of<br />

intestinal bacteria<br />

and hepatic coma.<br />

Neomycin Sulfate - - - - - ✔<br />

Suppression of<br />

bowel intestinal<br />

bacteria (e.g.<br />

preoperative<br />

preparation of the<br />

bowel); hepatic<br />

coma.<br />

22


Drug Infections (General) Gram Negative<br />

Infections<br />

Streptomycin Sulfate<br />

✔<br />

✔<br />

Use only in infections caused by Gram-negative bacilli<br />

organisms shown to be<br />

(in bacteremia, with<br />

susceptible, and when less<br />

another agent).<br />

potentially hazardous therapeutic<br />

agents are ineffective or<br />

contraindicated. Organisms<br />

usually sensitive include:<br />

Pasteurella pestis (plague);<br />

Francisella tularensis (tularemia);<br />

Brucella, Calymmatobacterium<br />

granulomatis (donovanosis,<br />

granuloma inguinale);<br />

Haemophilus ducreyi (chancroid);<br />

H. influenzae (in respiratory,<br />

endocardial and meningeal<br />

infections with another agent); K,<br />

pneumoniae pneumonia (with<br />

another agent); E. coli, Proteus<br />

sp., A. aerogenes, K. pneumoniae<br />

and Entercoccus faecalis in<br />

urinary tract infections;<br />

Streptococcus viridans and E.<br />

faecalis (in endocardial infections<br />

with penicillin).<br />

Staphylococcal Infections Neonatal Sepsis Intrathecal<br />

Administration<br />

Other<br />

- - - ✔<br />

Mycobacterium<br />

tuberculosis: The<br />

Advisory Council for<br />

the Elimination of<br />

TB, the American<br />

Thoracic Society and<br />

the CDC recommend<br />

either streptomycin or<br />

ethambutol be added<br />

as a fourth drug in a<br />

regimen containing<br />

isoniazid, rifampin<br />

and pyrazinamide for<br />

initial treatment of<br />

TB unless the<br />

likelihood of INH or<br />

rifampin resistance is<br />

very low. Reassess<br />

the need for a fourth<br />

drug when<br />

susceptibility testing<br />

results are known.<br />

Streptomycin is also<br />

indicated for therapy<br />

of TB when one or<br />

more of the above<br />

drugs is<br />

contraindicated<br />

because of toxicity or<br />

intolerance.<br />

23


Drug Infections (General) Gram Negative<br />

Infections<br />

Tobramycin Sulfate<br />

✔<br />

✔<br />

Treatment of serious bacterial In patients where serious<br />

infections caused by<br />

life-threatening gramnegative<br />

susceptible strains of<br />

infection is<br />

Pseudomonas aeruginosa, suspected, including<br />

Escherichia coli, Proteus sp.<br />

those in whom<br />

(indole-positive and indolenegative)<br />

concurrent therapy with<br />

including P.<br />

a penicillin or<br />

mirabilis, Morganella<br />

cephalosporin and an<br />

morganii and P. vulgaris, aminoglycoside may be<br />

Providencia sp. including<br />

indicated, initiate<br />

Klebsiella-Enterobacter-<br />

tobramycin before<br />

Serratia group, Citrobacter susceptibility study<br />

sp. and staphylococci<br />

results are obtained.<br />

including S. aureus<br />

Base decision to<br />

(coagulase-positive and<br />

continue therapy on<br />

coagulase-negative).<br />

these results.<br />

Infections including:<br />

Septicemia (neonates,<br />

children, adults, lower<br />

respiratory tract infections,<br />

serious CNS infections<br />

(meningitis), intra-abdominal<br />

infections, including<br />

peritonitis,<br />

skin, bone and skin structure<br />

infections,<br />

complicated and recurrent<br />

urinary tract infections<br />

(UTIs).<br />

Staphylococcal Infections Neonatal Sepsis Intrathecal<br />

Administration<br />

✔<br />

Tobramycin may be considered<br />

in serious staphylococcal<br />

infections when penicillin or<br />

other potentially less toxic drugs<br />

are contraindicated and when<br />

bacterial susceptibility testing<br />

and clinical judgment indicate its<br />

use.<br />

Other<br />

✔ - ✔<br />

Cystic Fibrosis<br />

(nebulizer only)<br />

24


IV.<br />

Pharmacokinetic Parameters of the Aminoglycoside Antibiotics<br />

Absorption<br />

Aminoglycosides are poorly absorbed from the GI tract, but are well absorbed following<br />

parenteral administration. 2 Intramuscular administration can result in interpatient variations in<br />

plasma concentrations achieved with a specific IM dose, due to differences in rates of absorption<br />

from injection sites. Aminoglycosides are also rapidly and almost completely absorbed following<br />

topical administration during surgical procedures or from surgical sites.<br />

Distribution<br />

Following absorption, aminoglycosides are widely distributed into body fluids, primarily in the<br />

extracellular fluid volume. Streptomycin appears to be 35% bound to plasma proteins, where<br />

other aminoglycosides are only minimally protein bound. 2 Additionally, a small portion of each<br />

aminoglycoside does accumulate in body tissues and is tightly bound intracellularly. Most body<br />

compartments and tissues including the inner ear and kidneys become progressively saturated with<br />

an aminoglycoside over the course of therapy, and the drug is slowly released from these areas.<br />

This may account for the ototoxicity and nephrotoxicity associated with these agents. The agents<br />

in this class differ in their affinity for renal tissue; streptomycin has less affinity for renal tissue<br />

than the other aminoglycosides.<br />

Elimination<br />

The plasma elimination half-life of aminoglycosides is usually 2-4 hours in adults with normal<br />

renal function. 2 Plasma concentrations are higher and plasma elimination half-lives are more<br />

prolonged in patients with impaired renal function. These concentrations are not usually affected<br />

by hepatic impairment, however, the plasma elimination half-life of streptomycin has been<br />

reported to be more prolonged in patients with both renal and hepatic impairment than in patients<br />

with renal impairment alone. In infants, aminoglycoside plasma elimination half-lives are<br />

inversely proportional to birthweight and gestational age and reflect renal maturity.<br />

Aminoglycosides are not metabolized and are excreted unchanged in urine by glomerular<br />

filtration. With normal renal function, 40-97% of a single IM or IV dose of an aminoglycoside is<br />

excreted in the urine within 24 hours. Complete recovery of a dose in the urine, in a patient with<br />

normal renal function requires approximately 10-20 days. Aminoglycosides are readily removed<br />

by hemodialysis and to a lesser extent by peritoneal dialysis. Table 5 illustrates the<br />

pharmacokinetic parameters of the aminoglycosides.<br />

Table 5. Pharmacokinetic Parameters of the Aminoglycosides 2,6<br />

Various Pharmacokinetic Parameters of the Aminoglycosides<br />

Therapeutic Toxic serum<br />

Half-life (hrs) serum levels levels(mcg/ml) Dose<br />

Aminoglycoside Normal ESRD (peak) Peak 1 Trough 2 (mg/kg/day)<br />

(mcg/ml)<br />

(normal Ccr)<br />

Amikacin 2-3 24-60 16-32 > 35 > 10 15<br />

Gentamicin 2 24-60 4-8 > 12 > 2 3-5<br />

Kanamycin 2-3 24-60 15-40 > 35 > 10 15<br />

Neomycin 2-3 12-24 - - - -<br />

Streptomycin 2.5 100 20-30 > 50 -- 15<br />

Tobramycin 2-2.5 24-60 4-8 > 12 > 2 3-5<br />

1 Measured 1 hour after IM administration.<br />

2 Measured immediately prior to next dose.<br />

25


V. Drug Interactions<br />

Neurotoxic, ototoxic, or nephrotoxic effects may be additive, concurrent and/or sequential with use<br />

of an aminoglycoside and other drugs with similar toxic potentials (e.g. other aminoglycosides,<br />

acyclovir, amphotericin B, bacitracin, capreomycin, cephalosporins, colistin, cisplatin,<br />

methoxyflurane, polymyxin B, vancomycin) should be avoided, if possible. 2 In addition, due to<br />

increased risk of ototoxicity, from additive effects or altered serum and tissue concentrations,<br />

aminoglycosides should not be given concurrently with ethacrynic acid, furosemide, urea, or<br />

mannitol.<br />

Concurrent use of aminoglycosides with general anesthetics or neuromuscular blocking agents may<br />

potentiate neuromuscular blockage and cause respiratory paralysis. When used together, patients<br />

should be observed for signs of respiratory depression.<br />

Oral neomycin may potentiate the effects of oral anticoagulants, thereby interfering with GI<br />

absorption or synthesis of vitamin K. 2 Prothrombin times should be monitored and the dose of the<br />

anticoagulant adjusted as required. Oral neomycin has been reported to decrease GI absorption of<br />

digoxin and methotrexate. Serum digoxin concentrations should be monitored when oral neomycin<br />

therapy is initiated or discontinued in patients previously stabilized on digoxin. Oral neomycin is<br />

also reported to decrease the rate but not the extent of absorption of spironolactone.<br />

In vitro studies indicate that the antibacterial activity of aminoglycosides, as mentioned previously,<br />

and β-lactam antibiotics or vancomycin may be additive or synergistic against some organisms<br />

including enterococci and Ps. aeruginosa. 2 Studies also indicate that aminoglycosides and<br />

extended-spectrum penicillins also exert a synergistic bacterial effect against some<br />

enterobacteriaceae. Serum aminoglycoside concentrations should be monitored in patients<br />

receiving concomitant therapy, especially when very high doses of the penicillin are used or if the<br />

patient has impaired renal function. Additionally, chloramphenicol, clindamycin, and tetracycline<br />

have been reported to antagonize the bactericidal activity of aminoglycosides.<br />

Indomethacin has been reported to increase trough and peak serum aminoglycoside concentrations<br />

in premature neonates who were receiving the drugs together. 2 Increases in serum aminoglycoside<br />

concentrations appeared to be related to indomethacin-induced decreases in urine output. It also<br />

has been proposed that inhibitors of prostaglandin synthesis (e.g. aspirin) may increase<br />

nephrotoxicity of aminoglycosides. Monitoring is important in premature neonates who are<br />

receiving an aminoglycoside and indomethacin. Table 6 describes the level 1 and level 2 (the most<br />

severe) drug-drug interactions with the aminoglycosides.<br />

26


Table 5. Drug Interactions of the Aminoglycosides 7<br />

Drug Significance Interaction Mechanism<br />

Aminoglycosides Level 1 Aminoglycosides and<br />

nondepolarizing muscle<br />

relaxants<br />

Aminoglycosides Level 1 Aminoglycosides and loop<br />

diuretics<br />

Aminoglycosides Level 2 Aminoglycosides and<br />

NSAIDs<br />

Aminoglycosides Level 2 Aminoglycosides and<br />

penicillins<br />

Aminoglycosides Level 2 Aminoglycosides and<br />

succinylcholine<br />

Aminoglycosides Level 2 Aminoglycosides and<br />

cephalosporins<br />

Aminoglycosides Level 2 Aminoglycosides and<br />

digoxin<br />

Possible pharmacologic synergism. Actions of the nondepolarizing<br />

muscle relaxants may be enhanced.<br />

Mechanism is not exactly known. But possible synergistic<br />

auditory toxicity is suspected. Auditory toxicity may be<br />

increased. Hearing loss of varying degrees may occur.<br />

Irreversible hearing loss has occurred.<br />

NSAIDs may cause an accumulation of aminoglycosides by<br />

reducing the glomerular filtration rate. Plasma aminoglycoside<br />

concentrations may be elevated.<br />

Mechanism is unknown. Certain parenteral penicillins may<br />

inactivate certain aminoglycosides. Do not mix parenteral<br />

aminoglycosides and penicillins in the same solution.<br />

Aminoglycosides may be additive or synergistic with<br />

succinylcholine secondary to their ability to stabilize the<br />

postjunctional membrane and impair prejunctional calcium influx<br />

and acetylcholine output. The resulting effect is that<br />

aminoglycosides can potentiate the neuromuscular effects of<br />

succinylcholine.<br />

Mechanism is unknown. Nephrotoxicity may be increased.<br />

Bactericidal activity against certain pathogens may be enhanced.<br />

The mechanism for the reduction in rate and extent of absorption is<br />

unknown. The potential for decreased metabolism is presumed to<br />

be caused by neomycin’s killing of intestinal bacteria that contribute<br />

to the formation of digoxin-reduction products. The rate and extent<br />

of digoxin absorption may be reduced, which could reduce the<br />

pharmacologic effect of the drug. However, in a small number of<br />

patients (15g of the drug should be observed carefully for signs of eighth cranial nerve<br />

damage. Loss of hearing has occurred with kanamycin, even with normal renal function.<br />

Aminoglycosides should be used with extreme caution in patients with neuromuscular disorders<br />

such as myasthenia gravis or parkinsonian syndrome, since these drugs can aggravate muscle<br />

weakness and result in a potential to produce neuromuscular blockade.<br />

Table 7 reports the common adverse events, by percentage, for the aminoglycosides.<br />

27


Table 7. Common Adverse Events (%) Reported for the Aminoglycosides 6<br />

Central/Peripheral Nervous<br />

System<br />

Adverse Reaction Amikacin Gentamicin Kanamycin Neomycin Streptomycin Tobramycin<br />

Headache rare 1 rare<br />

Encephalopathy<br />

Confusion<br />

Fever<br />

Lethargy<br />

Disorientation<br />

Neuromuscular blockade<br />

Paresthesia rare rare<br />

Convulsions<br />

Muscle twitching<br />

Myasthenia gravis-like syndrome<br />

Numbness<br />

Peripheral neuropathy<br />

Skin tingling<br />

Vomiting rare rare<br />

GI<br />

Nausea rare rare<br />

Diarrhea<br />

rare<br />

Anemia<br />

Eosinophilia<br />

rare<br />

rare<br />

Hematologic<br />

Leukopenia<br />

Thrombocytopenia<br />

Granulocytopenia<br />

Rash rare rare<br />

Hypersensitivity<br />

Lab test<br />

abnormalities<br />

Renal 1<br />

Urticaria<br />

Itching<br />

Anaphylaxis/Anaphylactoid<br />

reaction<br />

Increased AST/ALT<br />

Increased bilirubin<br />

Increased serum LDH<br />

Oliguria<br />

Proteinuria<br />

Rising serum creatinine<br />

Casts<br />

Nephrotoxicity<br />

Rising BUN<br />

Red and white cells in urine<br />

Azotemia<br />

28


Rising NPN<br />

Decreasing Ccr<br />

Dizziness<br />

Tinnitus<br />

Vertigo<br />

Special senses<br />

Roaring in ears<br />

Hearing loss/deafness 2<br />

Ototoxicity<br />

Loss of balance 2<br />

Visual disturbances/blurred vision<br />

Apnea<br />

Miscellaneous<br />

Drug fever rare rare<br />

Pain/Irritation at injection site<br />

Hypotension<br />

rare<br />

Acute muscular paralysis<br />

Decreased serum Ca, Na, K, Mg<br />

Malabsorption syndrome (incr. fecal<br />

fat)<br />

C. difficile associated colitis<br />

1<br />

= Reported; no incidence given.<br />

2 Partially reversible to irreversible bilateral hearing loss.<br />

VII.<br />

Dosing and Administration for the Aminoglycoside Antibiotics<br />

Current clinical evidence suggests that once-daily administration of aminoglycosides is at least as<br />

effective as, and may be less toxic than, conventional dosage regimens employing multiple daily<br />

doses of the drugs. 2 Further controlled studies are needed in children, patients with renal<br />

impairment, and other special groups, to determine optimal use.<br />

Because therapeutic and toxic concentrations of the aminoglycosides may be narrow, peak and<br />

trough concentrations should be determined periodically, especially in patients with renal<br />

impairment.<br />

Table 8. Dosing for the Aminoglycoside Agents 5-6<br />

Drug Availability Dose /Frequency/Duration<br />

Amikacin Sulfate Injection: 50mg/ml<br />

(pediatric), 250mg/ml<br />

Ideal body weight should be used for dosage calculation.<br />

Injection for IV infusion:<br />

500mg ADD-Vantage system<br />

and 500mg in 0.9% sodium<br />

chloride<br />

Administer IM or IV at 15mg/kg/day divided into 2 or 3 equal doses at<br />

equally divided intervals, over 30-60 minutes. Dosing in heavier patients<br />

should not exceed 1.5g/day. In uncomplicated UTIs, use 250mg twice<br />

daily.<br />

Neonates: Loading dose of 10mg/kg, followed by 7.5mg/kg every<br />

12 hours. Preliminary IM studies in newborns of different weights (<<br />

1.5kg, 1.5 to 2kg, > 2kg) at a dose of 7.5mg/kg revealed that, like other<br />

aminoglycosides, serum half-life values were correlated inversely with<br />

postnatal age and renal clearances of amikacin. Lower dosages may be<br />

safer during the first 2 weeks of life.<br />

29


Gentamicin Sulfate<br />

Injection: 10mg/ml<br />

(pediatric), 40mg/ml<br />

Injection for IV infusion:<br />

60, 80, or 100mg ADD-<br />

Vantage system, 40, 60, 70,<br />

80, 90, 100, and 120mg in<br />

0.9% sodium chloride<br />

Duration: Usual duration of treatment is 7 to 10 days. Do not exceed<br />

15mg/kg/day. For treatment beyond 10 days, monitor amikacin serum<br />

levels and renal, auditory and vestibular functions daily. Uncomplicated<br />

infections caused by sensitive organisms should respond in 24 to 48<br />

hours. If definite clinical response does not occur within 3 to 5 days, stop<br />

therapy and reevaluate. Failure of the infection to respond may be<br />

because of resistance of the organism or to the presence of septic foci<br />

requiring surgical drainage. Peak and trough concentrations should be<br />

monitored.<br />

May be given IM or IV.<br />

For serious infections and normal renal function, give 3mg/kg/day in 3<br />

equal doses every 8 hours. For life-threatening infections, administer up<br />

to 5mg/kg/day in 3 or 4 equal doses. Reduce dosage to 3mg/kg/day as<br />

soon as clinically indicated.<br />

Obese patients: Base dosage on an estimate of lean body mass.<br />

Children: 6 to 7.5mg/kg/day (2 to 2.5mg/kg every 8 hours).<br />

Infants and neonates: 7.5mg/kg/day (2.5mg/kg every 8 hours).<br />

Premature or full term neonates (≤1 week of age):<br />

5mg/kg/day (2.5mg/kg every 12 hours). A regimen of either<br />

2.5mg/kg every 18 hours or 3mg/kg every 24 hours may also<br />

provide satisfactory peak and trough levels in preterm infants < 32<br />

weeks gestational age.<br />

Prevention of bacterial endocarditis:<br />

(In dental, oral or upper respiratory tract procedures (alternate regimen)) 9 :<br />

1 to 2g (50mg/kg for children) ampicillin plus 1.5mg/kg (2mg/kg<br />

for children) gentamicin not to exceed 80mg, both IM or IV onehalf<br />

hour prior to procedure, followed by 1.5g (25mg/kg for<br />

children) amoxicillin 6 hours after initial dose or repeat parenteral<br />

dose 8 hours after initial dose.<br />

GU or GI procedures (standard regimen):<br />

2g (50mg/kg for children) ampicillin plus 1.5mg/kg (2mg/kg for<br />

children)gentamicin not to exceed 80mg, both IM or IV one-half<br />

hour prior to procedure followed by 1.5mg (25mg/kg for children)<br />

amoxicillin.<br />

Intrathecal: Administer only the 2 mg/ml intrathecal preparation without<br />

preservatives. Dosage will vary depending upon factors such as age and<br />

weight of the patient, site of injection, degree of obstruction to CSF flow<br />

and the amount of CSF estimated to be present.<br />

Duration: Usually 7 to 10 days. In difficult and complicated infections,<br />

a longer course of therapy may be necessary. In such cases, monitor<br />

renal, auditory and vestibular function, since toxicity is more apt to occur<br />

with treatment extended beyond 10 days. Reduce dosage if clinically<br />

indicated.<br />

30


Kanamycin Sulfate<br />

Neomycin Sulfate<br />

Oral: 500mg capsules<br />

Injection: 37.5/ml<br />

(pediatric), 250mg/ml, and<br />

333mg/ml<br />

Oral: 500mg tablets,<br />

125mg/5ml oral solution<br />

Oral:<br />

Suppression of intestinal bacteria<br />

As an adjunct to mechanical cleansing of the large bowel in short-term<br />

therapy - 1g every hour for 4 hours, followed by 1g every 6 hours for 36<br />

to 72 hours.<br />

Hepatic coma:<br />

8 to 12g/day in divided doses.<br />

Injection: Based on ideal body weight.<br />

Do not exceed a total of 1.5 g/day by any route.<br />

IM:<br />

Inject deeply into the upper outer quadrant of the gluteal muscle. For<br />

adults or children, 7.5mg/kg every 12 hours (15mg/kg/day). If<br />

continuously high blood levels are desired, give the daily dose of<br />

15mg/kg in equally divided doses every 6 or 8 hours. Usual treatment<br />

duration is 7 to 10 days. Doses of 7.5mg/kg give mean peak levels of<br />

22mcg/ml. At 8 hours after a 7.5mg/kg dose, mean serum levels are<br />

3.2mcg/ml.<br />

IV:<br />

Adults:<br />

Do not exceed 15mg/kg/day. Give slowly. Prepare by adding<br />

contents of 500mg vial to 100 to 200ml sterile diluent (Normal<br />

Saline or 5% Dextrose in Water) or the contents of a 1g vial to 200<br />

to 400ml of sterile diluent. Give over 30 to 60 minutes. Divide<br />

daily dose into 2 to 3 equal doses.<br />

Children:<br />

Use sufficient diluent to infuse the drug over 30 to 60 minutes.<br />

Other routes:<br />

Intraperitoneal: 500 mg diluted in 20ml sterile distilled water instilled<br />

through a polyethylene catheter into the wound.<br />

Aerosol: 250 mg 2 to 4 times a day. Withdraw 250 mg (1 ml) from<br />

500mg vial, dilute with 3ml Normal Saline and nebulize.<br />

Hepatic coma:<br />

Adults:<br />

4 to 12g/day in divided doses for 5-6 days.<br />

Children:<br />

50 to 100mg/kg/day in divided doses. Continue treatment over a<br />

period of 5 to 6 days; during this time, return protein to the diet<br />

incrementally. Chronic hepatic insufficiency may require up to<br />

4g/day over an indefinite period.<br />

Preoperative intestinal antisepsis:<br />

Neomycin is given for 24 hours, not to exceed 72 hours, at a usual adult<br />

dose of 1g each hour for 4 doses, then 1g every 4 hours for the balance of<br />

the 24 hours. A low residue diet should be prescribed, and a saline<br />

cathartic should be given immediately preceding initiation of neomycin.<br />

For the 2-3 day regimen, the usual daily dose for adults and children is<br />

88mg/kg in 6 equally divided doses at 4 hour intervals. Alternatively, it<br />

has been recommended that the following regimen be used for scheduled<br />

surgery at 8:00am: 1g neomycin and 1g erythromycin base administered<br />

at 1pm, 2pm, and 11pm on the day preceding surgery, as an adjunct to<br />

mechanical cleansing of the intestine.<br />

31


Streptomycin<br />

Sulfate<br />

Injection: 1g (200mg/ml<br />

Lyophilized cake/powder)<br />

Injection for IM use:<br />

400mg/ml<br />

Administer by the IM route.<br />

Tuberculosis:<br />

The standard regimen for the treatment of drug-susceptible TB has been 2<br />

months of INH, rifampin and pyrazinamide followed by 4 months of INH<br />

and rifampin (patients with concomitant TB and HIV infection may<br />

require treatment for a longer period). When streptomycin is added to<br />

this regimen because of suspected or proven drug resistance, the<br />

recommended dosing for streptomycin is as follows:<br />

Streptomycin Dosing for TB<br />

Daily Twice weekly Thrice weekly<br />

Children<br />

Adults<br />

20-40mg/kg<br />

max 1g<br />

15mg/kg<br />

max 1g<br />

25-30mg/kg<br />

max 1.5g<br />

25-30mg/kg<br />

max 1.5g<br />

25-30mg/kg<br />

max 1.5g<br />

25-30mg/kg<br />

max 1.5g<br />

Streptomycin is usually administered daily as a single IM injection. Give<br />

a total dose of 120 g over the course of therapy unless there are no other<br />

therapeutic options. In patients > 60 years of age, use a reduced dosage.<br />

Therapy with streptomycin may be terminated when toxic symptoms have<br />

appeared, when impending toxicity is feared, when organisms become<br />

resistant, or when full treatment effect has been obtained. The total period<br />

of drug treatment of TB is a minimum of 1 year.<br />

Tularemia:<br />

1 to 2g daily in divided doses for 7 to 14 days until the patient is afebrile<br />

for 5 to 7 days.<br />

Plague:<br />

2g daily in two divided doses for minimum of 10 days.<br />

Bacterial endocarditis:<br />

Streptococcal:<br />

Streptomycin may be used for 2 week treatment concomitantly<br />

with penicillin in penicillin-sensitive alpha and non-hemolytic<br />

streptococcal endocarditis (penicillin MIC 0.1 mcg/ml): 1g twice<br />

daily for 1 week, 0.5g twice daily for the second week. If patient is<br />

> 60 years of age, give 0.5g twice daily for the entire 2 week<br />

period.<br />

Enterococcal:<br />

1g twice daily for 2 weeks and 0.5g twice daily for 4 weeks in<br />

combination with penicillin. Ototoxicity may require termination<br />

of streptomycin prior to completion of the 6-week course of<br />

treatment.<br />

Concomitant agents:<br />

For use with other agents to which infecting organism is also sensitive,<br />

streptomycin is a secondary choice for treatment of gram-negative<br />

bacillary bacteremia, meningitis and pneumonia; brucellosis; granuloma<br />

inguinale; chancroid; UTI.<br />

Adults:<br />

1 to 2g in divided doses every 6 to 12 hours for moderate to severe<br />

infections. Doses should generally not exceed 2g per day.<br />

Children:<br />

20 to 40mg/kg/day (8 to 20mg/lb/day) in divided doses every 6 to 12<br />

hours. (Take particular care to avoid excessive dosage in children.)<br />

32


Tobramycin<br />

Sulfate<br />

Oral Inhalation for<br />

Nebulization: 300mg/5ml<br />

Injection: 10mg/ml<br />

(pediatric), 40mg/ml<br />

Injection for IV infusion:<br />

1.2g bulk package, 60 or<br />

80mg Add-Vantage system,<br />

60 and 80mg in 0.9% sodium<br />

chloride<br />

Use the patient's ideal body weight for dosage calculation; for obese<br />

patients, use patient's estimated lean body weight plus 40% of the excess as<br />

the basic weight on which to calculate mg/kg dosing.<br />

Adults:<br />

Administer 3mg/kg/day IV or IM in three equal doses every<br />

8 hours for serious infections or 5mg/kg/day IV or IM in three or<br />

four equal doses for life-threatening infections reduced to<br />

3mg/kg/day as soon as clinically indicated. To prevent increased<br />

toxicity caused by excessive blood levels, do not exceed<br />

5mg/kg/day, unless serum levels are monitored.<br />

Children:<br />

Administer 6 to 7.5mg/kg/day in 3 or 4 equally divided doses (2 to<br />

2.5mg/kg every 8 hours or 1.5 to 1.9mg/kg every 6 hours).<br />

Premature or full-term neonates (≤1 week of age):<br />

Administer 4mg/kg/day in 2 equal doses every 12 hours.<br />

Preliminary data suggest that 2.5mg/kg every 18 hours or 3mg/kg<br />

every 24 hours may achieve safe and effective peak and trough<br />

serum concentrations in newborn infants weighing 6 years of age and older the dose is 300mg Q12hours<br />

for 28 days.<br />

Duration:<br />

Usual duration of treatment is 7 to 10 days. A longer course may<br />

be necessary in difficult and complicated infections. In such cases,<br />

monitor renal, auditory and vestibular functions; toxicity can occur<br />

when treatment extends > 10 days.<br />

33


Special Dosing Considerations<br />

2, 5, 6<br />

Table 9. Special Dosing Considerations for the Aminoglycoside Antibiotics<br />

Drug Renal Hepatic Pediatric Use Pregnancy<br />

Amikacin<br />

Sulfate<br />

Gentamicin<br />

Sulfate<br />

Kanamycin<br />

Sulfate<br />

Neomycin<br />

Sulfate<br />

Streptomycin<br />

Sulfate<br />

Tobramycin<br />

Sulfate<br />

Dosing<br />

Yes<br />

Dosing<br />

and/or<br />

frequency<br />

Yes<br />

Dosing<br />

and/or<br />

frequency<br />

Yes<br />

Adjust<br />

frequency<br />

Dosing<br />

No<br />

No<br />

No<br />

Yes<br />

Neonates and<br />

infants<br />

Yes<br />

Neonates and<br />

infants<br />

Yes<br />

Children and<br />

infants<br />

Yes No Children only (no<br />

specific age limits<br />

found)<br />

Yes<br />

Dosing<br />

and/or<br />

frequency<br />

Yes<br />

Dosing<br />

and/or<br />

frequency<br />

No<br />

No<br />

Children only (no<br />

specific age limits<br />

found)<br />

Yes<br />

Children and<br />

premature or fullterm<br />

infants<br />

Category<br />

C<br />

C<br />

D<br />

C<br />

D<br />

C<br />

Stability<br />

Amikacin is stable for 24 hours at room<br />

temperature at concentrations of 0.25 and<br />

5mg/ml in 0.9% sodium chloride and 5%<br />

dextrose. Amikacin products are also stable<br />

for 24 hours at room temperature at the same<br />

concentrations after being refrigerated at 4<br />

degrees C for 60 days or frozen for 30 days<br />

at –15 degrees C, thawed, and stored at 25<br />

degrees C. Amikacin should not be mixed<br />

with other drugs.<br />

Gentamicin sulfate is stable for 24 hours at<br />

room temperature in most IV infusion fluids<br />

including 0.9% sodium chloride and 5%<br />

dextrose. These mixtures should not be<br />

mixed with other drugs.<br />

Kanamycin is stable for 24 hours at room<br />

temperature in most IV infusion fluids<br />

including 0.9% sodium chloride and 5%<br />

dextrose. The drug should not be mixed<br />

with other drugs.<br />

Neomycin oral solution should be stored in<br />

tight, light-resistant containers, at<br />

15-30 degrees C.<br />

Sterile reconstituted solutions of<br />

streptomycin sulfate are stable for one week<br />

when stored at room temperature and<br />

protected from light, however, streptomycin<br />

sulfate powder for injection contains no<br />

preservatives and the possibility of microbial<br />

contamination of reconstituted solutions<br />

must be considered.<br />

Following reconstitution of the sterile<br />

powder, tobramycin solutions containing<br />

40mg/ml are stable for 24 hours at room<br />

temperature or 96 hours at 2-8 degrees C.<br />

Tobramycin sulfate is stable for 24 hours at<br />

room temperature in most IV infusion<br />

solutions, however, the drug is incompatible<br />

with IV solutions containing alcohol.<br />

Commercially available tobramycin solution<br />

for oral inhalation should be stored at<br />

2-8 degrees C. When refrigeration is not<br />

available, intact or opened foil pouches<br />

containing ampules of the solution may be<br />

stored at room temperature up to 25 degrees<br />

C for up to 28 days.<br />

34


VIII. Comparative Effectiveness of the Aminoglycoside Antibiotics<br />

The aminoglycosides have been available for use for many years. Streptomycin and kanamycin<br />

are the oldest drugs in the class. Recent clinical data largely focuses on resistance patterns with<br />

the aminoglycosides. Other clinical evidence has evaluated once-daily administration versus<br />

multiple daily dosing. Clinical support for the aminoglycosides is presented in Table 9.<br />

Table 10. Additional Outcomes Evidence for the Aminoglycosides<br />

Study Sample Study/Treatment<br />

/ Duration<br />

Ioannidis DG,<br />

et al. 10<br />

Metaanalysis<br />

of<br />

24 studies<br />

published<br />

between<br />

1991 and<br />

2003<br />

Variable therapy<br />

duration; drugs<br />

evaluated were<br />

amikacin (9<br />

studies), gentamicin<br />

(11), tobramycin<br />

(2), netilmicin (2),<br />

and tobramycin or<br />

netilmicin (1).<br />

Results<br />

In evaluating the efficacy and toxicity of multiple daily dosing (MDD) and<br />

once-daily dosing (ODD) for the aminoglycoside class, 24 studies in<br />

children were evaluated and showed:<br />

Efficacy<br />

• There was no significant difference between ODD and MDD in<br />

the clinical failure rate, microbiologic failure rate, and combined<br />

clinical or microbiologic failure rates, but trends favored ODD<br />

consistently.<br />

• A statistically significant benefit was seen with ODD over MDD<br />

in trials using amikacin, whereas no statistical difference was seen<br />

in trials using other antibiotics.<br />

Nephrotoxicity<br />

• There was no significant difference between ODD and MDD in<br />

the primary nephrotoxicity outcomes. Secondary nephrotoxicity<br />

outcomes were significantly better with ODD.<br />

Ototoxicity<br />

• There was no significant difference between ODD and MDD in<br />

the primary ototoxicity outcomes.<br />

• Studies noting only the clinical impression of hearing impairment<br />

also failed to identify any toxicity (ODD: 114 cases; MDD:114<br />

cases).<br />

Subgroup and Bias Analysis (Summary)<br />

• No statistically significant differences between ODD and MDD<br />

in any of the examined subgroups (neonatal intensive care unit,<br />

cystic fibrosis, cancer, or urinary tract infection), with respect to<br />

combined clinical or microbiologic failure outcomes, primary<br />

nephrotoxic outcomes, or ototoxicity, when sufficient data were<br />

available.<br />

• Clinical failures were uncommon in the pediatric trials, and if<br />

anything, fewer clinical failures tended to occur with ODD.<br />

• There was no evidence to show any reduction in the risk of<br />

primary nephrotoxicity outcomes with ODD. However, the event<br />

rate was much lower among children, compared with adults, and<br />

the secondary nephrotoxicity outcomes favored ODD.<br />

• Although single trials have been small, the available randomized<br />

evidence supports the general adoption of ODD with<br />

aminoglycosides in pediatric clinical practice. Advantages of<br />

ODD include: simplification of administration, similar or<br />

potentially improved efficacy and safety, compared with MDD.<br />

Shawar RM,<br />

et al. 11 n=508 - In vitro activity of tobramycin was compared with those of six other<br />

antimicrobial agents against 1,240 Pseudomonas aeruginosa isolates<br />

collected from 508 patients with cystic fibrosis during pretreatment visits:<br />

• Tobramycin was the most active drug tested and also showed<br />

good activity against isolates resistant to multiple antibiotics.<br />

• The isolates were less frequently resistant to tobramycin<br />

(5.4%) than to ceftazidime (11.1%), aztreonam (11.9%), amikacin<br />

(13.1%), ticarcillin (16.7%), gentamicin (19.3%), or ciprofloxacin<br />

(20.7%).<br />

35


Mithani H, et<br />

al. 12 n=200 100 consecutive<br />

patients treated with<br />

QD dosing were<br />

evaluated via<br />

retrospective chart<br />

review, while 100<br />

consecutive patients<br />

treated with<br />

conventional<br />

regimens over the<br />

same calendar<br />

period were<br />

evaluated.<br />

• Of 56 isolates for which the tobramycin minimum inhibitory<br />

concentration (MIC) was > or = 16mcg/ml and that were<br />

investigated for resistance mechanisms, only seven (12.5%)<br />

were shown to possess known aminoglycoside-modifying<br />

enzymes; the remaining were presumably resistant by an<br />

incompletely understood mechanism often referred to as<br />

"impermeability."<br />

This retrospective analysis was designed to compare once-daily with<br />

conventional aminoglycoside administration, in terms of efficacy and<br />

toxicity.<br />

• Although this was a pharmacoeconomic analysis, only<br />

clinical efficacy data will be shared in this review.<br />

• Eighty-nine patients were cured or improved with once-daily<br />

administration versus 90 patients with conventional<br />

administration.<br />

• One patient in each group developed definite aminoglycosideinduced<br />

renal toxicity.<br />

• Summary: Once-daily administration of aminoglycosides is<br />

as effective and well tolerated, compared with<br />

aminoglycoside treatment utilizing conventional regimens.<br />

King JW, et<br />

al. 13 - - This prospective study was done because of growing resistance to<br />

gentamicin and tobramycin in isolates of aerobic and facultative gramnegative<br />

bacteria. The study tracked resistance, bacteremic episodes, and<br />

bacteremia-associated deaths before and after institution of amikacin as the<br />

sole preferred aminoglycoside:<br />

• From June 1984-June 1987 (before amikacin became<br />

preferred), resistance to gentamicin, tobramycin, and<br />

amikacin among aerobic and facultative gram-negative<br />

bacterial isolates were 12.8%, 10.8%, and 5.9%, respectively.<br />

• Once amikacin became preferred, over the next 30 months,<br />

the rates of resistance to gentamicin, tobramycin, and<br />

amikacin were 6.3%, 5.0%, and 3.3%, respectively.<br />

• Also, during the 30 months with amikacin was preferred, the<br />

incidence of both bacteremia and bacteremia-associated death<br />

decreased significantly.<br />

• Summary: Hospitals where resistance to gentamicin or<br />

tobramycin is increasing among gram-negative flora, may<br />

benefit from use of amikacin as the principal aminoglycoside.<br />

Gerding DN,<br />

et al. 14 - - For 10 years, the 700 bed Minneapolis Veterans Affairs Medical Center in<br />

Minnesota has monitored and carefully controlled aminoglycoside usage and<br />

resistance of over 25,000 aerobic and faculatative gram-negative bacillary<br />

isolates to the aminoglycosides. A summary of their findings includes:<br />

• Introduction of amikacin at a high level of usage in the 1980’s was<br />

associated with a significant reduction in resistance to gentamicin<br />

and tobramycin among gram-negative bacilli.<br />

• Rapid introduction of gentamicin usage in 1982 after use of<br />

amikacin was associated with a significant and rapid increase in<br />

gentamicin and tobramycin resistance. However, in 1986,<br />

gentamicin was again reintroduced to the institution and the usage<br />

of gentamicin was gradually increased over a 15-month period<br />

without significant change in resistance to gentamicin, tobramycin,<br />

or amikacin.<br />

• A move to a new hospital in June 1988 was associated with an<br />

additional significant decline in resistance to all aminoglycosides<br />

(p


Muscato JJ, et<br />

al. 15 - 3-month period<br />

when amikacin was<br />

restricted and<br />

tobramycin and<br />

gentamicin were<br />

unrestricted<br />

(baseline), followed<br />

by a period of 12<br />

months when<br />

amikacin was the<br />

primary<br />

aminoglycoside.<br />

Evans DA, et<br />

al. 16 - Qualitative<br />

overview of<br />

randomized trials<br />

comparing two or<br />

more of the<br />

following:<br />

amikacin,<br />

gentamicin,<br />

netilmicin,<br />

sisomicin, and<br />

tobramycin<br />

Lewis RT. 17 n=215 Double-blind,<br />

placebo-controlled,<br />

randomized trial of<br />

combined oral and<br />

systemic antibiotics<br />

vs. systemic<br />

antibiotics; also a<br />

meta-analysis of<br />

randomized studies<br />

comparing<br />

combined versus<br />

systemic antibiotics<br />

in elective colon<br />

surgery<br />

This study in 12 cancer treatment centers in the United States, was designed<br />

to evaluate the potential for increased resistance to amikacin with<br />

unrestricted use:<br />

• Amikacin usage increased from a mean of 20.1% to a mean<br />

of 83.9% of aminoglycoside patient-days.<br />

• A reduction in the use of tobramycin and gentamicin were<br />

observed with means of 66.1 and 10%, and 13.9 and 6.1%,<br />

respectively for the two periods.<br />

• Resistance to amikacin was 0.85% at baseline and 1.3% at<br />

end-point, which was not clinically significant (p=0.614).<br />

• Baseline resistance was 6.5 and 7.6%, while final resistance<br />

was 2.6 and 4.8%, respectively, for tobramycin (p=0.001) and<br />

gentamicin (p=0.052).<br />

In evaluating randomized trials comparing aminoglycosides with respect to<br />

efficacy, nephrotoxicity, and auditory toxicity:<br />

• The results of most trials showed no significant differences,<br />

although sample sizes were, in general, smaller than<br />

necessary to detect moderate differences.<br />

• Quantitative overviews may be a useful means of detecting<br />

real differences in risk that are not apparent from the results<br />

of several individual small trials.<br />

In comparing the efficacy of combined oral (neomycin and metronidazole)<br />

and systemic antibiotics (amikacin and metronidazole) versus the same<br />

systemic antibiotics alone in preventing surgical site infection:<br />

• Wound infections occurred in 5 patients in the combined<br />

group but in 17 of the systemic group (p < 0.01, RR = 0.29,<br />

95% CI 0.11-0.75).<br />

• Bacteria isolated from wound infections and wound fat were<br />

similar to those found in the colon. They were more frequent<br />

in the colon in the systemic group (p < 0.001) and occurred in<br />

wound fat in the systemic group twice as often as in the<br />

combined group (p < 0.001).<br />

• By stepwise logistic regression, the presence of bacteria in<br />

wound fat at surgery was the strongest predictor of<br />

postoperative wound infection (p < 0.002).<br />

In the meta-analysis (of studies from 1975-1995) of randomized studies<br />

comparing combined versus systemic antibiotics, studies indicated:<br />

• The summary weighted risk difference in surgical site infections<br />

between groups and the summary risk ratios both favored<br />

combined prophylaxis = 0.56, 95% CI 0.26-0.86; RR = 0.51, 95%<br />

CI 0.24-0.78; p < 0.001).<br />

• Summary: In elective surgery of the colon, combined oral and<br />

systemic antibiotics are superior to systemic antibiotics in<br />

preventing surgical site infections. Orally administered antibiotics<br />

add value by reducing bacterial load of the colon and would fat<br />

contamination, both associated with postoperative wound<br />

infection.<br />

Song F, et<br />

al. 18 - 147 relevant<br />

randomized,<br />

controlled trials<br />

were identified in a<br />

systematic review<br />

To access the relative efficacy of antimicrobial prophylaxis for the<br />

prevention of postoperative wound infection in patients undergoing<br />

colorectal surgery:<br />

• The results confirm that the use of antimicrobial prophylaxis is<br />

effective for the prevention of surgical wound infection after<br />

colorectal surgery.<br />

• There was no significant difference in the rate of surgical wound<br />

infections between many different regimens.<br />

• However, certain regimens appear to be inadequate (e.g.<br />

metronidazole alone, doxycycline alone, piperacillin alone, oral<br />

neomycin plus erythromycin on the day before operation).<br />

37


Mitch WE, et<br />

al. 19 n=14 5-7 days before and<br />

during oral<br />

neomycin or<br />

kanamycin<br />

• A single dose administered immediately before the operation (or<br />

short-term use) is as effective as long-term postoperative<br />

antimicrobial prophylaxis (odds ratio 1.17 (95% confidence<br />

interval 0.90-1.53)).<br />

• There is no convincing evidence to suggest that the newgeneration<br />

cephalosporins are more effective than first-generation<br />

cephalosporins (odds ratio 1.07 (95% CI 0.54-2.12)).<br />

• Summary: Antibiotics selected for prophylaxis in colorectal<br />

surgery should be active against both aerobic and anaerobic<br />

bacteria (e.g. aminoglycosides plus metronidazole).<br />

Administration should be timed to make sure that the tissue<br />

concentration of antibiotics around the wound area is sufficiently<br />

high when bacterial contamination occurs.<br />

In patients with severe chronic renal failure who received long-term<br />

nutritional therapy with protein restriction and supplements of amino acids<br />

or their nitrogen free analogues, nitrogen balance was studied during<br />

administration of neomycin or kanamycin:<br />

• There was a significant improvement in nitrogen balance<br />

during the antibiotic period when compared to the control<br />

period, averaging +0.80g of nitrogen per day (p


(92%) in the SD group (Mantel-Haenszel odds ratio 0.36; CI 95%,<br />

1.19-0.64; p = 0.0004).<br />

• Summary: In human brucellosis the treatment of rifampicin and<br />

doxycycline presents a greater number of recurrence and lower<br />

number of cure than the classical treatment with streptomycin and<br />

tetracycline drugs.<br />

Additional Evidence<br />

Dose Simplification: Clinical data has been presented above on once-daily versus multiple daily<br />

dosing of the aminoglycosides. Single daily dosing of the aminoglycosides is possible because of<br />

their rapid concentration-dependent killing and post-antibiotic effect and has the potential for<br />

decreased toxicity. Single daily dosing appears to be safe and efficacious, although, in some<br />

clinical situations (endocarditis and pediatric use), traditional multiple dosing is still usually<br />

recommended. 1 There was no significant difference between ODD and MDD in the clinical failure rate,<br />

microbiologic failure rate, and combined clinical or microbiologic failure rates.<br />

Stable Therapy: Stable therapy is of particular interest to all classes of antibiotic use, including<br />

aminoglycosides, as changing therapies can lead to drug resistance.<br />

In the interest of preferred drug list management, little scientific research is available on<br />

antimicrobial switching. Antimicrobial switching can be used as a mechanism to reduce or<br />

prevent antimicrobial resistance. This is often the case with nosocomial gram-negative organisms<br />

that can rapidly develop resistance. 4 The Centers for Disease Control is currently researching<br />

antimicrobial switch programs in an effort to better define resistance patterns within a therapy<br />

class caused by overuse of a particular agent. Research is also looking at whether discontinuation<br />

of an agent in a class can restore susceptibility.<br />

Some institutions have formed an antibiotic subcommittee to join local representatives from<br />

microbiology, infection control, pharmacy, infectious disease, and multiple disciplines of medical<br />

practice, in an effort to monitor resistance patterns and recommend treatment guidelines.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal specific<br />

clinical data on the impact of medical utilization and use of the aminoglycosides. Medical<br />

resources utilized are dependent on the severity of the infection and the condition of the patient.<br />

IX.<br />

Conclusions<br />

Efficacy of the aminoglycosides is dependent on the target indication for use and resistance<br />

patterns, or susceptibility of the identified organism to the drugs in this class. Gentamicin,<br />

tobramycin, and amikacin are considered similar in efficacy for the treatment of gram-negative<br />

organisms including Ps. aeruginosa. Gentamicin is the most commonly used aminoglycoside.<br />

Amikacin is likely the aminoglycoside of choice when gentamicin resistance is strongly<br />

suspected. 22 Neomycin use is limited mainly to oral therapy for surgical bowel preparation or<br />

hepatic coma. Streptomycin has only a few specific indications, as newer agents are available that<br />

have broader spectrums of activity. Kanamycin is indicated in serious gram-negative infections in<br />

which Ps. aeruginosa is not a likely causative agent.<br />

There is at least one available generic formulation for each aminoglycoside reviewed in this class.<br />

As a result, all brand products within the class reviewed are comparable to each other and to the<br />

generics and OTC products in this class and offer no significant clinical advantage over other<br />

alternatives in general use.<br />

39


Recommendations<br />

No brand aminoglycoside is recommended for preferred status.<br />

40


References<br />

1. Gonzalez LS III, Spencer JP. Aminoglycosides: A Practical <strong>Review</strong>. Am Fam Physician 1998<br />

Nov 15;58(8):1811-20.<br />

2. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

3. Chambers HF, Sande MA. The aminoglycosides. In: Hardman JG, Limbird LE, eds. Goodman<br />

and Gilman’s The pharmacological basis of therapeutics. 9 th ed. New York: McGraw-Hill,<br />

1996;1103-21.<br />

4. Abate BJ, Barriere SL. Antimicrobial Regimen Selection. In: <strong>Pharmacotherapy</strong>. A<br />

Pathophysiologic Approach, Fifth Edition. Dipiro JT, Talbert RL, Yee GC, et al. Eds. McGraw-<br />

Hill. New York. 2002. Pg. 1817-29.<br />

5. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58,<br />

2004. Thomson PDR. Montvale, NJ. 2004.<br />

6. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

7. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

8. Briggs GG, Freeman RK, Yaffe SJ. Drugs in pregnancy and lactation. A reference guide to fetal<br />

and neonatal risk. Sixth Edition. Lippincott, Williams, & Wilkins, Philadelphia, 2002.<br />

9. Dajani AS, Bisno AL, Chung KJ, et al. Prevention of bacterial endocarditis. Recommendations<br />

by the American Heart Association. JAMA1990;264:2919-2922.<br />

10. Contopoulos-Ioannidia DG, Giotis ND, Baliatsa DV, et al. Extended-interval aminoglycoside<br />

administration for children: a meta-analysis. Pediatrics 2004 Jul;114(1):e111-8.<br />

11. Shawar RM, MacLeod DL, Garber RL, et al. Activities of tobramycin and six other antibiotics<br />

against Pseudomonas aeruginosa isolates from patients with cystic fibrosis. Antimicrob Agents<br />

Chemother 1999 Dec;43(12):2877-80.<br />

12. Mithani H, Brown G. The economic impact of once-daily versus conventional administration of<br />

gentamicin and tobramycin. Pharmacoeconomics 1996 Nov;10(5):494-503.<br />

13. King JW, White MC, Todd JR, et al. Alterations in the microbial flora and in the incidence of<br />

bacteremia at a university hospital after adoption of amikacin as the sole formulary<br />

aminoglycoside. Clin Infect Dis 1992 Apr;14(4):908-15.<br />

14. Gerding DN, Larson TA, Hughes RA, et al. Aminoglycoside resistance and aminoglycoside<br />

usage: ten years of experience in one hospital. Antimicrob Agents Chemother 1991<br />

Jul;35(7):1284-90.<br />

15. Muscato JJ, Wilbur DW, Stout JJ, et al. An evaluation of the susceptibility patterns of gramnegative<br />

organisms isolated in cancer centers with aminoglycoside usage. J Antimicrob<br />

Chemother 1991 May;27 Suppl C:1-7.<br />

16. Evans DA, Buring J, Mayrent S, et al. Qualitative overview of randomized trials of<br />

aminoglycosides. Am J Med 1986 Jun30;80(6B):39-43.<br />

17. Lewis RT. Oral versus systemic antibiotic prophylaxis in elective colon surgery: a randomized<br />

study and meta-analysis send a message from the 1990s. Can J Surg 2002 Jun;45(3):173-80.<br />

18. Song F, Glenny AM. Antimicrobial prophylaxis in colorectal surgery: a systematic review of<br />

randomized controlled trials. Br J Surg 1998 Sep;85(9):1232-41.<br />

19. Mitch WE, Walser M. Effects of oral neomycin and kanamycin in chronic uremic patients: II.<br />

Nitrogen balance. Kidney Int 1977 Feb;11(2):123-7.<br />

20. Boulanger LL, Ettestad P, Fogarty JD, et al. Gentamicin and tetracyclines for the treatment of<br />

human plague: review of 75 cases in New Mexico, 1985-1999. Clin Infect Dis 2004 Mar<br />

1;38(5):663-9.<br />

21. Solera J, Martinez-Alfaro E, Saez L. Meta-analysis of the efficacy of the combination of rifampin<br />

and doxycycline in the treatment of human brucellosis. Med Clin (Barc) 1994 May<br />

21;102(19):731-8.<br />

22. Brewer NS. Antimicrobial agents—Part II. The aminoglycosides: streptomycin, kanamycin,<br />

gentamicin, tobramycin, amikacin, neomycin. Mayo Clin Proc 1977 Nov;52(11):675-9.<br />

41


I. Overview<br />

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Antifungal Agents<br />

AHFS 081400<br />

October 27, 2004<br />

There are three major types of complications caused by the approximately 200 known species of<br />

fungi that infect humans. These complications are allergies, mycotoxicoses and mycoses. A few<br />

dozen fungi are the etiology for 90% of mycoses. The incidence of superficial and systemic<br />

mycotic infections has increased dramatically over the past decade. The two major factors<br />

contributing to this shift in epidemiology are related to two specific immunocompromised patient<br />

populations: patients undergoing transplantation and chemotherapy and patients with HIV and/or<br />

AIDS. In response to the increasing incidence of fungal infections, several oral and injectable<br />

agents that are effective against systemic fungal pathogens have been developed. 1-17<br />

The availability over the past two decades of the azole antifungal agents represents a major<br />

advance in the management of systemic fungal infections. The relative broad spectrum of the<br />

azoles against common fungal pathogens (Candida sp., Cryptococcus neoformans, Blastomyces<br />

dermatitidis, Histoplasma capsulatum, Coccidioides immitis, Parcoccidioides brasiliensis,<br />

Sporothix schenckii and Aspergillus sp), ease of administration, and limited toxicity are highly<br />

attractive features. Fluconazole, itraconazole, ketoconazole, and voriconazole are azole<br />

antifungals that inhibit the CYT P450 dependent synthesis of ergosterol, the principal sterol in the<br />

fungal cell membrane, and are approved to treat systemic mycoses. Griseofulvin, an oral agent, is<br />

only effective in the treatment of superficial dermatophytes. Terbinafine is a synthetic allylamine<br />

derivative that inhibits squalene epoxidase, a key enzyme in sterol biosynthesis in fungi, resulting<br />

in a deficiency in ergosterol within the fungal cell membrane and cell death. Terbinafine is only<br />

approved to treat topical onychomycosis of the fingernails and toenails. Among the azoles,<br />

fluconazole possesses the most desirable pharmacologic properties, including high bioavailability,<br />

high water solubility, a low degree of protein binding, a wide volume of distribution into body<br />

tissues and fluids including cerebrospinal fluid, and urine, and long half-life. In addition,<br />

fluconazole and itraconazole are better tolerated and more effective than ketoconazole.<br />

Voriconazole is an azole antifungal agent derived from the structure of fluconazole. It was<br />

designed to enhance the potency and spectrum of activity of fluconazole. In addition to the<br />

original indications (invasive aspergillosis, refractory infection due to Scedosporium sp. or<br />

Fusarium sp.) voriconazole has recently been approved for treatment of esophageal candidiasis.<br />

One potential limitation of the azole antifungals is the drug-drug interactions, which are more<br />

significant with itraconazole and ketoconazole.<br />

Though the incidence of mycosis has increased, there remains a limited population in which headto-head<br />

comparative trials for the most common fungi can be conducted. Hence, comparative<br />

efficacy data are limited. Studies are available for topical infections such as onychomycosis and<br />

systemic infections in the HIV population. It is also important to note that duration of treatment is<br />

based on the severity of the patient's underlying disease, level of immunosuppression, and clinical<br />

response. 1-17 Table 1 lists the agents included in this review. This review encompasses all<br />

dosage forms and strengths.<br />

42


Table 1. Antifungal Agents in this <strong>Review</strong><br />

Generic Name Antifungal Type Formulation Example Brand Name (s)<br />

Amphotericin B Polyene Injection *Fungizone ® , *Amphocin ®<br />

Amphotericin B Cholesteryl<br />

Polyene Injection Amphotec ® (ABCD)<br />

Sulfate Complex<br />

Amphotericin B Lipid<br />

Polyene Injection Abelcet ® (ABLC)<br />

Complex<br />

Amphotericin B Liposomal Polyene Injection AmBisome ®<br />

Caspofungin Acetate Echinocandin Injection Cancidas ®<br />

Fluconazole Azole Oral/ Injection *Diflucan ®<br />

Flucytosine Pyrimidine Oral Ancobon ®<br />

Itraconazole Azole Oral/ Injection Sporanox ®<br />

Ketoconazole Azole Oral *Nizoral ®<br />

Nystatin Polyene Oral *Mycostatin ®<br />

Terbinafine Allylamine Oral Lamisil ®<br />

Voriconazole Azole Oral / Injection Vfend ®<br />

Griseofulvin Microsize Misc. Antifungal Oral Fulvicin U/F ® , Grifulvin V ®<br />

Griseofulvin Ultramicrosize Misc. Antifungal Oral *Gris-PEG ®<br />

*Generic Available.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Treatment guidelines for the antifungal agents range from the treatment of opportunistic infections<br />

in patients with human immunodeficiency virus (HIV), to neutropenic patients with cancer,<br />

cryptococcemia, candidiasis, transplantation, and treatment of onychomycosis. For purposes of<br />

the scope of this review, treatment guidelines for onychomycosis and candidiasis will be briefly<br />

reviewed.<br />

Candidiasis<br />

Candida species are the most common cause of fungal infections. 18, 19 This species produces<br />

infections that range from non-life-threatening mucocutaneous illness to invasive, full-blown<br />

infections that may involve multiple organs. Due to the broad range of infections, an equally<br />

broad range of diagnostic and therapeutic strategies are necessary.<br />

43


Table 2. Guidelines for the Treatment of Candidiasis<br />

Recommendations<br />

• Knowledge of the infecting species is highly predictive of likely susceptibility and can be<br />

used as a guide to therapy.<br />

• Invasive Candidiasis: Extensive data from randomized trials is available only for therapy<br />

of acute hematogenous candidiasis in the nonneutropenic patient. Choice of therapy for<br />

other forms of candidiasis is based on case series and anecdotal reports. In general,<br />

amphotericin B-based agents, the azole antifungal agents, and the echinocandin antifungal<br />

agents play a large role in treatment. Choice of therapy is guided by weighing the greater<br />

activity of amphotericin B-based agents and the echinocandin antifungal agents for some<br />

non-albicans species (e.g. Candida krusei) against the availability of oral and parenteral<br />

formulations for the azole antifungal agents. Flucytosine has activity against many<br />

isolates of Candida but is infrequently used.<br />

Initial therapy usually involves caspofungin, fluconazole, amphotericin B, or a<br />

combination of fluconazole and amphotericin B. Experience with caspofungin in adults is<br />

still somewhat limited. Neonates with disseminated candidiasis are usually treated with<br />

amphotericin B, and fluconazole has been used successfully in small numbers of neonates.<br />

The guidelines comment in detail on specific susceptibilities with Candida species.<br />

• Mucocutaneous Candidiasis: Therapy for mucosal infections is dominated by the azole<br />

antifungal agents. These drugs can be used topically or systemically and are safe and<br />

efficacious. Some patients may have repeated relapses with possible induction of<br />

resistance with prolonged or repeated exposure. Initial episodes can be treated with<br />

clotrimazole troches or nystatin. Oral fluconazole is as effective as, and in some studies,<br />

superior to topical therapy. Itraconazole solution is as efficacious as fluconazole.<br />

Ketoconazole and itraconazole capsules are less effective than fluconazole, because of<br />

variable absorption. Systemic therapy is required for effective treatment of esophageal<br />

candidiasis.<br />

• Prevention of Invasive Candidiasis: Prophylactic strategies are useful in those<br />

populations where risk is high, such as patients undergoing therapy that produces<br />

prolonged neutropenia (bone marrow transplant or solid-organ transplant). For<br />

neutropenic patients, fluconazole or itraconazole is appropriate therapy for patients at a<br />

significant risk for invasive candidiasis. Institutions where high rates of invasive<br />

candidiasis in adult or neonatal ICU exist despite standard infection control procedure<br />

could consider fluconazole prophylaxis for carefully selected patients in these areas.<br />

Onychomycosis<br />

Onychomycosis accounts for one-third of fungal skin infections. Because only one-half of nail<br />

dystrophies are caused by fungus, diagnosis should be confirmed by potassium hydroxide<br />

preparation, culture, or histology prior to treatment being inititiated. 20 Terbinafine and<br />

itraconazole are the agents of choice for the treatment of onychomycosis, while fluconazole has<br />

not been specifically indicated for the treatment of onychomycosis, but early data has been<br />

promising in its use. Table 3 describes treatment guidelines for onychomycosis and Table 4<br />

discusses information highlighted in a Food and Drug Administration Public Health Advisory<br />

concerning safety of itraconazole and terbinafine for the treatment of onychomycosis.<br />

44


Table 3. Guidelines for the Treatment of Onychomycosis 21<br />

Recommendations<br />

• Complete a comprehensive history of presenting symptoms with attention to history of<br />

onset, duration, morphology of nail, predisposing factors, and prior treatments and<br />

outcomes.<br />

• Physician exam looking at degree of severity: nail color, texture, nail base angle, plate<br />

firmness and adherence to the nail bed. Severe onychomycosis is considered >80% of<br />

the nail surface and/or matrix involvement of at least one toenail (excluding the 5 th<br />

metatarsal).<br />

• Diagnostic tests: microscopy, Wood’s lamp examination, skin/nail biopsy, and fungal<br />

culture (if considering long-term oral therapy). Differential diagnosis include lichen<br />

planus, psoriasis, myxoid cyst, yellow nail syndrome, peripheral vascular disease, and<br />

trauma.<br />

• Lifestyle modifications may aid in the prevention of onychomycosis. Important<br />

modifications include: wearing properly fitting shoes, using shower shoes in community<br />

showers, washing feet daily with soap and water, wearing hosiery made of synthetic<br />

materials, and supplying manicurist with personal pedicure/manicure tools.<br />

• *Amorolfine (Loceryl) 5% applied once weekly for 24 weeks in combination with<br />

itraconazole (Sporanox) 200mg orally every day for 12 weeks is strongly suggested as<br />

the most effective means of mycological and clinical cure for severe non-dermatophyte<br />

onychomycosis. Amorolfine (Loceryl) 5% applied once weekly for 15 months in<br />

combination with terbinafine (Lamisil) 250mg orally every day for 12 weeks is strongly<br />

supported as the most effective means of mycological and clinical cure for severe<br />

dermatophyte onychomycosis. Amorolfine is not available in the United States at this<br />

time.<br />

• Use of 40% Urea (Carmol 40) is recommended when applied 1-2 times daily until<br />

complete chemical avulsion of the nail is achieved. This is indicated for moderate to<br />

severe nail involvement in patients >18 years of age. Some evidence suggests beneficial<br />

effects of 40% Urea as a preparatory agent to avulse the nail and leave the nail bed more<br />

permeable to treatment with a topical antifungal.<br />

• Oral antifungals as monotherapy have been strongly supported for the treatment of<br />

moderate to severe onychomycosis. Terbinafine 250mg orally every day for 6 weeks for<br />

fingernails and 12-16 weeks for toenails has been shown to be the most effective<br />

treatment of dermatophyte infection. Itraconazole 200mg administered orally every day<br />

for 12 weeks of pulse therapy (400mg PO QD x one week Q4 weeks for 12-16 weeks)<br />

has been shown to be the most effective treatment of non-dermatophyte infection, with<br />

greater efficacy in favor of the pulse therapy. Fluconazole 150mg once weekly for 24<br />

weeks demonstrated lower efficacy in clinical and mycological cure of non-dermatophyte<br />

infections.<br />

• Complete blood count and liver function tests should be monitored prior to, during, and at<br />

the end of treatment with oral antifungals. When using nail lacquer treatments, nails<br />

should be cut and filed monthly by a healthcare professional.<br />

• Referral should be made to a podiatrist for severe and relapsing cases.<br />

*Amorolfine is not available in the United States at this time.<br />

45


FDA Public Health Advisory<br />

In April 2001, the FDA issued a Public Health Advisory for itraconazole and terbinafine for the<br />

use of onychomycosis. 22 The advisory came after the announcement of safety-related updates to<br />

the labeling of itraconazole and terbinafine tablets.<br />

The FDA advised that physicians not prescribe itraconazole to treat fungal infections<br />

(onychomycosis) in patients who have congestive heart failure (CHF), or a history of CHF. The<br />

updated labeling also includes contraindications and precautions with certain medicines.<br />

The advisory alerted the public that itraconazole and terbinafine have been associated with serious<br />

liver problems including liver failure and death. The advisory did not pertain to terbinafine cream<br />

and solution. As a result of the advisory, the FDA recommended revisions to labeling for both<br />

drugs, indicating that healthcare professionals should obtain nail specimens for laboratory testing<br />

to confirm the diagnosis of onychomycosis, prior to prescribing these medications.<br />

Table 4. FDA Public Health Advisory: Use of Itraconazole and Terbinafine for Onychomycosis 22<br />

FDA Recommendation Highlights<br />

• Itraconazole Cardiac Risk: As of April 2001, the FDA received and reviewed 94 cases<br />

in which patient’s taking the drug developed congestive heart failure. In 58 of the cases,<br />

the FDA ruled the drug contributed to or may have been the cause of the CHF. Although<br />

the causal relationship is unclear, death was reported in 13 cases.<br />

• Itraconazole and Terbinafine Hepatic Risk: As of March 2001, the FDA reviewed 24<br />

cases of liver failure possibly associated with itraconazole, including 11 deaths. There<br />

were 16 reviewed cases for the same timeframe of terbinafine associated liver failure,<br />

including 11 deaths and two liver transplant patients.<br />

III.<br />

Comparative Indications of the Antifungal Agents<br />

The azole antifungal drugs are contraindicated in those patients who have a hypersensitivity to<br />

other azole derivatives. Terbinafine is contraindicated in patients with a hypersensitivity to<br />

terbinafine, naftifine or any component of the drug. The griseofulvin drugs are contraindicated in<br />

those with a hypersensitivity to griseofulvin or any component of the formulation, porphyria<br />

(interferes with porphyrin metabolism), and hepatocellular failure. Amphotericin B should not be<br />

used in patients with a hypersensitivity to the drug or any other component of the formulation<br />

unless the condition requiring treatment is life-threatening and amenable only to amphotericin B<br />

therapy. The lipid formulation amphotericin agents are contraindicated in patients with known<br />

hypersensitivity to any amphotericin B product or any other constituents of the product unless, if<br />

the benefits of therapy outweigh the risk.<br />

Additionally, both itraconazole and ketoconazole have black box warnings as part of their<br />

labeling. Details on each drug’s warnings are included in bold in Table 5.<br />

46


1-17, 23-25<br />

Table 5. FDA-Approved Indications for the Antifungal Agents<br />

Drug<br />

Indication<br />

Amphotericin B<br />

Deoxycholate<br />

Amphotericin B Cholesteryl<br />

Sulfate Complex<br />

Potentially life-threatening fungal infections including:<br />

• Aspergillosis<br />

• North American blastomycosis<br />

• Systemic candidiasis<br />

• Coccidioidomycosis<br />

• Cryptococcosis<br />

• Histoplasmosis<br />

• Paracoccidioidomycosis<br />

• Sporotrichosis<br />

• Zygomycosis<br />

• Protozoal infections (leishmaniasis and primary amebic<br />

meningoencephalitis caused by Naegleria fowleri)<br />

The drug also can be used for empiric antifungal therapy in febrile<br />

neutropenic patients and as secondary prophylaxis to prevent<br />

recurrence or relapse of fungal infections listed above.<br />

For the treatment of invasive aspergillosis in patients where renal<br />

impairment or unacceptable toxicity precludes the use of amphotericin<br />

B deoxycholate in effective doses, and in patients with invasive<br />

aspergillosis where prior amphotericin B deoxycholate therapy has<br />

failed.<br />

Amphotericin B Lipid<br />

Complex<br />

For the treatment of invasive fungal infections in patients who are<br />

refractory to or intolerant of conventional IV amphotericin B.<br />

Amphotericin B Liposomal • Empirical therapy for presumed fungal infections in febrile,<br />

neutropenic patients.<br />

• Treatment of Cryptococcal meningitis in HIV infected patients.<br />

• Treatment of patients with Aspergillosis species, Candida species,<br />

and/or Cryptococcus species refractory to amphotericin B<br />

deoxycholate, or in patients where renal impairment or<br />

unacceptable toxicity precludes the use of amphotericin B<br />

deoxycholate.<br />

• For the treatment of visceral leishmaniasis. In<br />

immunocompromised patients with visceral leishmaniasis treated<br />

with amphotericin B liposomal, relapse rates were high following<br />

initial clearance of parasites.<br />

Caspofungin Acetate • Candidemia and the following Candida infections: intraabdominal<br />

abscesses, peritonitis and pleural space infections. The<br />

drug has not been studied in endocarditis, osteomyelitis, and<br />

meningitis due to Candida.<br />

• Esophageal candidiasis.<br />

• Invasive aspergillosis in patients refractory to or intolerant of other<br />

therapies (e.g. amphotericin B including lipid formulations, and/or<br />

itraconazole). The drug has not been studied as initial therapy for<br />

invasive aspergillosis.<br />

Fluconazole • Vaginal candidiasis<br />

• Oropharyngeal and esophageal candidiasis<br />

• Cryptococcal meningitis<br />

Flucytosine<br />

Serious infections caused by susceptible strains of Candida and/or<br />

Cryptococcus.<br />

Itraconazole • Blastomycosis<br />

• Histoplasmosis<br />

• Aspergillosis<br />

• Onychomycosis of the toenail/fingernail<br />

47


Black Box Warning:<br />

CHF:<br />

Do not administer itraconazole for the treatment of onychomycosis in<br />

patients with evidence of ventricular dysfunction, such as CHF or a<br />

history of CHF. Discontinue if signs and symptoms of CHF occur<br />

during treatment. If signs and symptoms of CHF occur during<br />

treatment, reassess the continued use of itraconazole. When<br />

itraconazole was administered by IV to dogs and healthy human<br />

volunteers, negative inotropic effects were seen.<br />

Drug interactions:<br />

Coadministration of cisapride, pimozide, dofetilide, or quinidine with<br />

itraconazole is contraindicated. Itraconazole is a potent inhibitor of the<br />

cytochrome P450 3A4 isoenzyme system and may raise plasma<br />

concentrations of drugs metabolized by this pathway. Serious<br />

cardiovascular events, including QT prolongation, torsades de pointes,<br />

ventricular tachycardia, cardiac arrest, and/or sudden death have<br />

occurred in patients taking itraconazole concomitantly with cisapride,<br />

pimozide, or quinidine, which are inhibitors of the cytochrome P450<br />

3A4 system (see contraindications, warnings, and drug Interactions).<br />

Ketoconazole • Candidiasis<br />

• Chronic mucocutaneous candidiasis<br />

• Oral thrush<br />

• Blastomycosis<br />

• Coccidioidomycosis<br />

• Histoplasmosis<br />

• Chromomycosis<br />

• Paracoccidioidomycosis<br />

• Recalcitrant cutaneous dermatophyte infection<br />

Black Box Warning:<br />

When used orally, ketoconazole has been associated with hepatic<br />

toxicity, including some fatalities. Patients receiving this drug should<br />

be informed by the physician of the risk and should be closely<br />

monitored. See Warnings and Precautions sections.<br />

Coadministration of terfenadine with ketoconazole tablets is<br />

contraindicated. Rare cases of serious cardiovascular adverse events,<br />

including death, ventricular tachycardia and torsades de pointes have<br />

been observed in patients taking ketoconazole tablets concomitantly<br />

with terfenadine, due to increased terfenadine concentrations induced<br />

by ketoconazole tablets. See contraindications, warnings, and<br />

precautions sections.<br />

Pharmacokinetic data indicate that oral ketoconazole inhibits the<br />

metabolism of astemizole, resulting in elevated plasma levels of<br />

astemizole and its active metabolite desmethylastemizole, which may<br />

prolong QT intervals. Coadministration of astemizole with<br />

ketoconazole tablets is therefore contraindicated. See<br />

contraindications, warnings, and precautions sections.<br />

Coadministration of cisapride with ketoconazole is contraindicated.<br />

Serious cardiovascular adverse events including ventricular<br />

tachycardia, ventricular fibrillation and torsades de pointes have<br />

occurred in patients taking ketoconazole concomitantly with cisapride.<br />

See contraindications, warnings, and precautions sections.<br />

Nystatin • Nonesophageal membrane GI candidiasis (cutaneous and<br />

mucocutaneous).<br />

• Prevention of fungal infections (HIV, transplant, and cancer).<br />

Terbinafine • Onychomycosis of toenail/fingernail.<br />

48


Voriconazole • Invasive aspergillosis<br />

• Esophageal candidiasis<br />

• Serious fungal infections caused by Scedosporium apiospermum<br />

(asexual form of Pseudallescheria boydii) and Fusarium spp.<br />

including Fusarium solani, in patients intolerant of, or refractory<br />

to, other therapy.<br />

Griseofulvin Ultramicrosize<br />

Griseofulvin Microsize<br />

Use only in minor or trivial infections that will not respond to<br />

topical agents alone. Effective only against dermatophytes.<br />

• Tinea pedis, tinea cruris, tinea corporis, tinea barbae, tinea capitis,<br />

and tinea unguium due to:<br />

• Trichophyton rubrum<br />

a. Trichophyton mentagrophytes<br />

b. Trichophyton tonsurans<br />

• Epidermophyton floccosum<br />

• Microsporum canis<br />

• Microsporum audouini<br />

• Microsporum gypseum<br />

• Trichophyton interdigitalis<br />

c. Trichophyton verrsusom<br />

d. Trichophyton megninii<br />

• Trichophyton gallinae<br />

e. Trichophyton crateriform<br />

f. Trichophyton sulphureum<br />

g. Trichophyton schoenleinii<br />

IV.<br />

Pharmacokinetic Parameters of the Antifungal Agents<br />

Table 6 describes the pharmacokinetic parameters of the antifungal agents in this class, while<br />

Table 7 details the kinetic properties of the amphotericin B formulations.<br />

As there appears to be striking differences in the pharmacokinetics of the amphotericin B lipid<br />

formulations, the clinical efficacy of the three formulations has turned out to be identical. 26 For<br />

example, the volume of distribution for the amphotericin B lipid complex is reported to greatly<br />

exceed that for liposomal amphotericin B. Some of the kinetic differences have been observed in<br />

numerous studies investigating time-concentration profiles of lipid formulations of amphotericin B<br />

and have been ascribed to the different particle sizes and shapes of the three lipid formulations.<br />

The Infectious Diseases Society of America does not recommend a preferred lipid amphotericin B<br />

product of the three available formulations.<br />

In a comparative pharmacokinetic evaluation of conventional amphotericin B, liposomal<br />

amphotericin B (AmBisome), and amphotericin B colloidal dispersion (Amphotec), the authors<br />

developed a chromatographic method for the separate measurement of lipid-formulated<br />

amphotericin B and amphotericin B (non-lipid) which has been liberated from its lipid binding. 26<br />

The liberated fraction of amphotericin B is bound to proteins in the plasma because amphotericin<br />

B is insoluble in water. Different kinetic characteristics were found for the liposomal<br />

amphotericin B and the colloidal dispersion amphotericin B, in particular for their lipid-formulated<br />

fractions. Although the mechanisms of liberation of amphotericin B from lipid binding are poorly<br />

understood, data suggests that after infusion of liposomal amphotericin B or colloidal dispersion,<br />

significant amounts of amphotericin B are liberated from the lipid moiety during circulation in the<br />

plasma. The pharmacokinetic properties of the liberated fraction are rather similar for both of the<br />

different formulations and resemble those of conventional amphotericin B.<br />

From a pharmacodynamic point of view, the equivalence in clinical efficacy of the different<br />

formulations supports a model that considers the liberated fraction of amphotericin B to be the<br />

active form. 26<br />

49


The lipid amphotericin formulations offer better therapeutic index without the toxicity seen with<br />

conventional amphotericin B. AmBisome is the only true liposome; Abelcet has a ribbon-like<br />

structure, and Amphotec is composed of disc-like structures. All of the lipid formulations contain<br />

amphotericin B, but they differ in shape, size, reticuloendothelial clearance (Cmax), AUC and<br />

visceral diffusion. 27<br />

1-17, 23-25<br />

Table 6. Pharmacokinetic Parameters of the Antifungal Agents<br />

Drug Bioavailability Protein<br />

Binding<br />

Metabolism Active<br />

Metabolites<br />

Elimination<br />

Caspofungin Acetate - 97% Hepatic - Renal 41%,<br />

Fecal 35%<br />

Half-Life<br />

Biphasic:<br />

1 st =9-11<br />

hours /<br />

2 nd =40-50<br />

hours<br />

Fluconazole >90% 11-12% Hepatic No Renal 60-80% 30 hours<br />

Flucytosine 75-90% 2-4% (at<br />

conc. 2-<br />

55mcg/ml)<br />

Minimal<br />

amounts are<br />

metabolized<br />

Itraconazole 40-55% 99.8% Hepatic<br />

CYP3A4<br />

No Renal 75-<br />

90%;<br />

unabsorbed<br />

drug is<br />

excreted in<br />

feces<br />

Yes Fecal 3-18%,<br />

Renal 40%<br />

Ketoconazole 75% 99% Hepatic No Fecal 57%,<br />

Renal 13%<br />

Nystatin<br />

Terbinafine<br />

Poorly absorbed<br />

from GI tract and<br />

mucous membranes;<br />

no detectable blood<br />

levels are obtained<br />

40% (1 st pass<br />

metabolism)<br />

- - - Almost<br />

entirely fecal<br />

>99% Hepatic: CYP<br />

2C19, CYP2C9<br />

and CYP 3A4<br />

Voriconazole 96% 58% Hepatic:<br />

CYP2C19, CYP<br />

2C9, CYP3A4<br />

Griseofulvin Ultramicrosize<br />

Griseofulvin Microsize<br />

(20-50)<br />

2.5-6 hours<br />

21 hours<br />

Biphasic:<br />

1 st =2 hours<br />

/ 2 nd =8<br />

hours<br />

-<br />

No Renal 70% 36 hours<br />

No Hepatic 98%;<br />

Renal 2%<br />

- - Hepatic - Urine (


Table 7. Pharmacokinetic Parameters of the Amphotericin B Agents after Multiple Doses 23<br />

Drug<br />

Peak Conc.<br />

(micrograms/ml)<br />

AUC 0-24 Clearance<br />

(mL/h•kg)<br />

Amphotericin B<br />

Deoxycholate<br />

(given at<br />

0.6mg/kg/day for<br />

42 days a )<br />

Amphotericin B<br />

Cholesteryl Sulfate<br />

Complex*<br />

Amphotericin B<br />

Lipid Complex*<br />

(given at<br />

5mg/kg/day for 5-7<br />

days<br />

Amphotericin B<br />

Liposomal*<br />

(5mg/kg/day)<br />

1.1 ± 0.2 (n=5) 17.1 ± 5<br />

(n=5)<br />

38 ± 15<br />

(n=5)<br />

2.9 36 0.028<br />

(L/h/kg)<br />

Volume of<br />

Distribution<br />

(Vd area )<br />

(L/kg)<br />

Terminal<br />

Elimination<br />

Half-Life<br />

(hours)<br />

5 ± 2.8 (n=5) 91.1 ± 40.9<br />

(n=5)<br />

Amt. Excreted in<br />

Urine Over 24 hours<br />

after Last Dose (% of<br />

dose) d<br />

9.6 ± 2.5 (n=8)<br />

1.1 39 -<br />

436 ± 131 ± 57.7 173.4 ± 78<br />

1.7 ± 0.8 (n=10) b 14 ± 7<br />

(n=14) b, c 188.5<br />

(n=14) b, c (n=8) c (n=8) c<br />

83.0 ± 35.2 555 ±<br />

311<br />

0.9 ± 0.4 (n=8) c<br />

11 ± 6 0.10 ± 0.07 6.8 ± 2.1 -<br />

* The assay used to measure amphotericin B in the blood after administration does not distinguish amphotericin B that is complexed with the<br />

phospholipids of Abelcet or AmBisome or cholesteryl sulfate, from amphotericin B that is uncomplexed.<br />

a<br />

Data from patients with mucocutaneous leischmaniasis. Infusion rate was 0.25mg/kg/h.<br />

b Data from studies in patients with cytology proven cancer being treated with chemotherapy or neutropenic patients with presumed or proven<br />

fungal infection. Infusion rate was 2.5mg/kg/h.<br />

c Data from patients with mucocutaneous leischmaniasis. Infusion rate was 4mg/kg/h.<br />

d Percentage of dose excreted in 24 hours after last dose.<br />

V. Drug Interactions of the Antifungal Agents<br />

All Level 1 interactions are highlighted in Table 8, along with other common drug-drug<br />

interactions with the antifungal agents. Level 1 interactions are the most life-threatening, and are<br />

usually rapid or delayed in onset, of major severity, and documentation has been established, is<br />

probable, or suspected.<br />

The lipid amphotericin B formulations have not been formally studied with regards to drug-drug<br />

interactions. The drugs known to interact with conventional amphotericin B are likely to also<br />

interact with the lipid formulations of the drug. There are no significant clinical differences<br />

among the three lipid formulations with regards to drug-drug interactions. As with conventional<br />

amphotericin B, caution should be used when the lipid amphotericin B formulations are<br />

administered together with other nephrotoxic medications such as aminoglycosides and<br />

pentamidine.<br />

Additionally, some antifungals have contraindications due to significant drug-drug interactions:<br />

Drug Interaction Contraindications<br />

Itraconazole (Sporanox): The following drugs are contraindicated with concomitant use of<br />

23, 28<br />

itraconazole, due to increased plasma concentration of the additional drug. Contraindicated<br />

drugs include: oral midazolam, pimozide, quinidine, dofetilide, and triazolam. The HMG-CoA<br />

reductase inhibitors, lovastatin and simvastatin, are also contraindicated.<br />

Ketoconazole (Nizoral): Both dofetilide and oral triazolam are contraindicated for<br />

23, 28<br />

coadministration with ketoconazole. Use of midazolam and triazolam has resulted in elevated<br />

plasma concentrations of the latter drugs, and may potentiate and prolong hypnotic and sedative<br />

effects, especially with repeated dosing or chronic administration.<br />

51


Voriconazole (Vfend): Coadministration of voriconazole and the CYP 3A4 substrates (pimozide,<br />

quinidine) is contraindicated since increased plasma concentrations of these drugs can lead to QT<br />

prolongation and rare occurrences of torsade de pointes. Voriconazole should also not be used in<br />

combination with sirolimus. Voriconazole should not be given with rifampin, carbamazepine, and<br />

long-acting barbiturates since these drugs are likely to decrease plasma concentrations of<br />

voriconazole. Use of rifabutin and voriconazole is contraindicated because voriconazole<br />

significantly increases rifabutin plasma concentrations and rifabutin also significantly reduces<br />

voriconazole plasma concentrations. Finally, voriconazole should not be used together with the<br />

ergot alkaloids (ergotamine and dihydroergotamine) because the drug can increase the<br />

concentrations of the ergot alkaloids and may lead to ergotism.<br />

23, 28<br />

Table 8. Clinically Significant Drug Interactions<br />

Drug Significance Interaction Mechanism<br />

Amphotericin B No Level 1<br />

interactions<br />

Level 2<br />

Others:<br />

• Cyclosporine and<br />

amphotericin B<br />

• Pentamidine<br />

The exact mechanism is unknown. The nephrotoxic<br />

effects of cyclosporine appear to be increased. The<br />

interaction occurs a few days following addition of<br />

amphotericin B to cyclosporine therapy and it is<br />

unknown how soon renal dysfunction resolves once<br />

amphotericin has been discontinued.<br />

Acute, reversible renal failure has been reported in at<br />

least 4 four patients who received amphotericin B<br />

concomitantly with IV or IM pentamidine.<br />

Amphotericin B Lipid<br />

Formulations<br />

(Cholesteryl Sulfate<br />

Complex, Lipid<br />

Complex, and<br />

Liposomal)<br />

No Level 1<br />

interactions<br />

• Drugs affected by<br />

potassium depletion<br />

Others: • Antineoplastic drugs<br />

• Corticosteroids and<br />

corticotrophin<br />

• Cyclosporin and<br />

tacrolimus<br />

• Digoxin<br />

• Flucytosine<br />

• Skeletal Muscle<br />

Relaxants<br />

Conventional amphotericin B can induce<br />

hypokalemia, patients receiving cardiac glycosides<br />

may be predisposed to glycoside-induced<br />

cardiotoxicity and may enhance the effects of skeletal<br />

muscle relaxants.<br />

Enhancement of the potential for renal toxicity,<br />

bronchospasm, and hypotension.<br />

May potentiate hypokalemia leading to cardiac<br />

dysfunction.<br />

In a randomized trial, adults and children<br />

receiving cyclosporine or tacrolimus in addition<br />

to Amphotec had a significantly lower rate of<br />

renal toxicity (31%, 16/51), compared to the<br />

conventional amphotericin B patients receiving<br />

cyclosporine or tacrolimus (68%, 17/49).<br />

Concurrent use with amphotericin B may<br />

induce hypokalemia and potentiate digoxin<br />

toxicity.<br />

Concurrent use may increase the toxicity of<br />

flucytosine by increasing cellular uptake and/or<br />

impairing its excretion.<br />

Amphotericin B induced hypokalemia may<br />

enhance the curariform effect of drugs such as<br />

tubocurarine, due to hypokalemia.<br />

52


Caspofungin Acetate<br />

Does not inhibit any of<br />

the CYP 450 enzymes<br />

• Tacrolimus<br />

Caspofungin reduced the peak concentration of<br />

tacrolimus by 16%, the AUC by 20%, and 12-hour<br />

blood concentration by 26%.<br />

• Cyclosporine<br />

In two clinical trials, cyclosporine increased the AUC<br />

of caspofungin by approximately 35%. Caspofungin<br />

did not increase levels of cyclosporine.<br />

Fluconazole<br />

Inhibits the metabolic<br />

activity of CYP 2C9,<br />

CYP 2C19, and<br />

CYP3A4.<br />

Level 1<br />

Level 1<br />

• Rifampin<br />

Warfarin and fluconazole<br />

Vincristine/vinblastine and<br />

fluconazole<br />

When given concomitantly with caspofungin, studies<br />

have shown a 30% decrease in caspofungin trough<br />

levels. This suggests other inducers of drug<br />

clearance (efavirenz, nevirapine, phenytoin,<br />

dexamethasone, and carbamazepine) may result in<br />

clinically meaningful reductions in caspofungin<br />

concentrations.<br />

Inhibition of warfarin metabolism. The anticoagulant<br />

effect of warfarin may be increased.<br />

Possible inhibition of vinca alkaloid metabolism<br />

(CYP3A4) by fluconazole, and all azole antifungal<br />

agents.<br />

Others:<br />

• Rifampin and<br />

cimetidine<br />

Increase the metabolism of fluconazole.<br />

• Oral contraceptives<br />

Fluconazole may decrease the effect of oral<br />

contraceptives.<br />

• HMG-CoA reductase<br />

inhibitors<br />

Azoles may inhibit the first-pass metabolism of the<br />

HMG-CoA reductase inhibitors, resulting in<br />

increased side-effects and plasma levels of these<br />

drugs.<br />

Flucytosine<br />

Itraconazole<br />

Inhibits the metabolic<br />

Significance<br />

not reported<br />

Others:<br />

• Cyclosporine,<br />

phenytoin,<br />

sulfonylureas,<br />

warfarin, triazolam,<br />

alprazolam,<br />

midazolam,<br />

tacrolimus,<br />

nortriptyline, rifabutin,<br />

zidovudine, and<br />

saquinavir.<br />

• Hydrochlorothiazide<br />

Cytosine arabinoside and<br />

flucytosine<br />

• Drugs that impair<br />

glomerular filtration<br />

may prolong the<br />

biological half-life of<br />

flucytosine.<br />

• Antifungal synergism<br />

between flucytosine<br />

and polyene antibiotics<br />

(amphotericin B) has<br />

been reported.<br />

Fluconazole increases the plasma concentration of<br />

these drugs.<br />

Concomitant administration resulted in significant<br />

fluconazole levels.<br />

Cytosine is a cytostatic agent and has been reported<br />

to inactivate the antifungal activity of flucytosine by<br />

competitive inhibition.<br />

Level 1 Warfarin and itraconazole Inhibition of warfarin metabolism. The anticoagulant<br />

effect of warfarin may be increased.<br />

53


activity of CYP 2C9<br />

and CYP 3A4.<br />

Level 1<br />

Pimozide and itraconazole<br />

Azole antifungals, including itraconazole, may inhibit<br />

the metabolism of pimozide. The risk of lifethreatening<br />

cardiac arrhythmias may be increased.<br />

Level 1<br />

Vinblastine / vincristine and<br />

itraconazole<br />

Possible inhibition of vinca alkaloid metabolism<br />

(CYP3A4) by fluconazole, and all azole antifungal<br />

agents.<br />

Others:<br />

• H-2 blockers, antacids,<br />

sucralfate, and<br />

omeprazole decrease<br />

the absorption of<br />

itraconazole.<br />

Studies have shown that absorption of itraconazole is<br />

impaired when gastric acid production is decreased.<br />

• Digoxin,<br />

carbamazepine,<br />

rifabutin, busulfan,<br />

alprazolam, diazepam,<br />

dihydropyridines,<br />

atorvastatin,<br />

cyclosporine,<br />

tacrolimus, sirolimus,<br />

oral hypoglycemics,<br />

protease inhibitors,<br />

buspirone, and<br />

methylprednisolone.<br />

The plasma concentration of these drugs may be<br />

increased by itraconazole.<br />

• Carbamazepine,<br />

phenytoin,<br />

phenobarbital,<br />

isoniazid, rifampin,<br />

rifabutin, antacids, H-2<br />

antagonists, proton<br />

pump inhibitors,<br />

nevirapine.<br />

These drugs may decrease the plasma concentration<br />

of itraconazole.<br />

Ketoconazole<br />

Inhibits the metabolic<br />

activity of CYP 3A4,<br />

CYP2C19, CYP 2C9<br />

and CYP 1A2<br />

Level 1<br />

Level 1<br />

• Clarithromycin,<br />

erythromycin,<br />

indinavir, ritonavir.<br />

Pimozide and ketoconazole<br />

Warfarin and ketoconazole<br />

These drugs may increase the plasma concentration<br />

of itraconazole.<br />

Azole antifungals, including ketoconazole, may<br />

inhibit the metabolism of pimozide. The risk of lifethreatening<br />

cardiac arrhythmias may be increased.<br />

Inhibition of warfarin metabolism. The anticoagulant<br />

effect of warfarin may be increased.<br />

Level 1<br />

Pimozide and ketoconazole<br />

Azole antifungals, including ketoconazole, may<br />

inhibit the metabolism of pimozide. The risk of lifethreatening<br />

cardiac arrhythmias may be increased.<br />

Level 1<br />

Vinblastine / vincristine and<br />

ketoconazole<br />

Possible inhibition of vinca alkaloid metabolism<br />

(CYP3A4) by ketoconazole, and all azole antifungal<br />

agents.<br />

Level 1<br />

Dofetilide and ketoconazole<br />

Inhibition of the renal cation transport system which<br />

is responsible for dofetilide elimination, resulting in<br />

elevated dofetilide plasma concentrations.<br />

54


Others: • H-2 antagonists,<br />

antacids, sucralfate,<br />

and omeprazole<br />

• Isoniazid, rifampin,<br />

phenytoin, and<br />

Pphenobarbital<br />

• Tacrolimus,<br />

cyclosporine,<br />

methylprednisolone,<br />

phenytoin,<br />

sulfonylureas,<br />

warfarin,<br />

benzodiazepines,<br />

indinavir, and<br />

ritonavir.<br />

Nystatin - Since nystatin absorption is<br />

insignificant, it is unlikely drug<br />

interactions would occur with<br />

nystatin and other<br />

concomitantly administered<br />

drugs. No interactions are<br />

Terbinafine<br />

Inhibits the metabolic<br />

activity of CYP 2D6<br />

No Level 1<br />

Interactions<br />

Level 2<br />

reported for nystatin lozenges.<br />

Tricyclic antidepressants<br />

(desipramine, imipramine,<br />

nortriptyline) and terbinafine.<br />

Decreased absorption of ketoconazole.<br />

Increase the metabolism of ketoconazole.<br />

Ketoconazole increases the plasma concentration of<br />

these drugs.<br />

-<br />

Inhibition of tricyclic antidepressant metabolism by<br />

CYP 2D6, resulting in increased pharmacologic and<br />

toxic effects of the tricyclic antidepressants.<br />

Level 2<br />

Others:<br />

Cyclosporine and terbinafine<br />

• Beta-blockers<br />

• SSRIs, MAOIs, and<br />

TCAs<br />

• Rifampin<br />

• Cimetidine<br />

Terbinafine may increase cyclosporine metabolism<br />

causing decreased cyclosporine concentrations.<br />

Terbinafine increases the clearance of cyclosporine<br />

by 15%.<br />

Decreased efficacy of beta-blockers.<br />

Increased toxicity of MAOIs, SSRIs, and TCAs.<br />

Rifampin increases terbinafine clearance by 100%.<br />

Cimetidine decreases terbinafine by 33%.<br />

Voriconazole<br />

Inhibits metabolic<br />

activity of CYP 2C19,<br />

CYP 2C9, and CYP<br />

• No data is available on<br />

terbinafine when given<br />

with oral<br />

contraceptives,<br />

hormone replacement<br />

therapy,<br />

hypoglycemics,<br />

theophyllines,<br />

phenytoins, thiazide<br />

diuretics, and calcium<br />

channel blockers.<br />

Level 1 Rifamycins and voriconazole Rifamycins increase metabolism of voriconazole and<br />

voriconazole inhibits the metabolism of rifabutin.<br />

55


3A4 Level 1<br />

Level 1<br />

Level 1<br />

Level 1<br />

Level 1<br />

Level 1<br />

Others:<br />

Griseofulvin No Level 1<br />

interactions.<br />

Level 2<br />

Level 2<br />

Level 2<br />

Others:<br />

Pimozide and voriconazole<br />

Barbiturates (Phenobarbital) and<br />

voriconazole<br />

Ergot Derivatives and<br />

voriconazole<br />

Carbamazepine and<br />

voriconazole<br />

Quinidine and voriconazole<br />

Warfarin and voriconazole<br />

Cyclosporine, phenytoin,<br />

sulfonylureas, triazolam,<br />

alprazolam, midazolam,<br />

loratadine, felodipine,<br />

nifedipine, statins, omeprazole,<br />

and tacrolimus.<br />

Barbiturates (Phenobarbital) and<br />

griseofulvin<br />

Warfarin and griseofulvin<br />

Oral contraceptives and<br />

griseofulvin<br />

• Cyclosporine and<br />

salicylates<br />

• Ethanol<br />

Voriconazole may inhibit the metabolism of<br />

pimozide, causing increased risk of life-threatening<br />

arrhythmias.<br />

Long-acting barbiturates may increase the<br />

metabolism of voriconazole, decreasing the<br />

therapeutic effect.<br />

Voriconazole may inhibit the metabolism of ergot<br />

derivatives causing increased risk of ergot toxicity.<br />

Carbamazepine may increase the metabolism of<br />

voriconazole, causing a decreased therapeutic effect<br />

of voriconazole.<br />

Voriconazole may inhibit the metabolism of<br />

quinidine, causing risk of life-threatening cardiac<br />

arrhythmias.<br />

Inhibition of warfarin metabolism. The anticoagulant<br />

effect of warfarin may be increased.<br />

Voriconazole increases the plasma concentration of<br />

these drugs.<br />

Decreased griseofulvin absorption and increased<br />

hepatic metabolism by phenobarbital result in<br />

decreased serum levels of griseofulvin.<br />

Mechanism is unknown. The anticoagulant activity<br />

of warfarin may be decreased.<br />

Griseofulvin induction of oral contraceptive hepatic<br />

metabolism is suspected, possibly leading to loss of<br />

oral contraceptive efficacy.<br />

Griseofulvin may decrease the efficacy of these<br />

drugs.<br />

Ethanol may increase griseofulvin toxicity, resulting<br />

in tachycardia and flushing.<br />

VI.<br />

Adverse Drug Events of the Antifungal Agents<br />

The most common adverse reactions with the amphotericin B products are infusion related chills<br />

23, 24<br />

and/or fever. Although full doses of amphotericin B may be tolerated by some patients, most<br />

will exhibit some intolerance, which can be improved with prophylactic treatment with aspirin,<br />

acetaminophen, antihistamines, or antiemetics. In a study designed to evaluate the incidence of<br />

infusion reactions occurring in patients receiving conventional amphotericin B, 71% of patients<br />

had at least one infusion-related reaction during the first seven days of therapy; fever and chills<br />

occurred in 28-51% and nausea and headache occurred in 9-18% of patients. 24 Additionally,<br />

meperidine has been shown to decrease the duration of shaking chills and fever that may<br />

accompany the infusion. Some benefit can be achieved with intravenous administration of small<br />

doses of adrenal corticosteroids just prior to or during the amphotericin B infusion, to decrease<br />

56


febrile reactions. Infusion related adverse reactions with Amphotec occur most frequently with<br />

the first dose of the drug (35% vs. 14% by the seventh dose). Tables 9 and 10 describe common<br />

adverse events with all of the antifungal drugs.<br />

Table 9. Common Adverse Events (%), by System, Reported for the Antifungal Agents 23<br />

Adverse Event<br />

Body as a Whole<br />

Chest Pain<br />

Generalized Pain<br />

Hypertriglyceridemia<br />

Cardiovascular<br />

Edema<br />

Hemorrhage<br />

Hypotension<br />

Hypertension<br />

Tachycardia<br />

Digestive System<br />

Abdominal Pain<br />

Nausea<br />

Vomiting<br />

Diarrhea<br />

Dyspepsia<br />

Appetite decrease<br />

Taste disturbance<br />

Flatulence<br />

Cramping Epigastric Pain<br />

Central Nervous System<br />

Chills/shaking<br />

Dizziness<br />

Fatigue<br />

Fever<br />

Headache<br />

Hallucinations<br />

Malaise<br />

Vertigo<br />

Insomnia<br />

Emotional Lability<br />

Nervousness<br />

Depression<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Cholestatic jaundice<br />

Hepatitis<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Hematologic<br />

Neutropenia<br />

Anemia<br />

Thrombocytopenia<br />

Renal<br />

Abnormal kidney function<br />

Acute kidney failure<br />

Other<br />

Allergic reaction<br />

Decreased libido<br />

Visual disturbance<br />

Weight Loss<br />

Injection Site Pain/Rxn<br />

Face Edema<br />

Weight Gain<br />

Multiple Organ Failure<br />

Sepsis<br />

Amphotericin<br />

B*<br />

11.6<br />

b, 12.8<br />

b<br />

b, 21.5<br />

16.3<br />

20.9<br />

21.8<br />

b, 38.7<br />

b, 43.9<br />

b, 27.3<br />

b<br />

b<br />

b, 75.9<br />

b<br />

b, 20.9<br />

b<br />

14.2<br />

ALT 14<br />

AST 12.8<br />

24.4<br />

10.2<br />

b, 42.2<br />

(creatinine<br />

increase)<br />

b<br />

b<br />

11.3<br />

Cholesteryl Sulfate<br />

(Amphotec)<br />

>5<br />

>5<br />

>5<br />

>5<br />

>5<br />

1-5<br />

>5<br />

>5<br />

b<br />

35<br />

>5<br />

35<br />

1-5<br />

>5<br />

1-5<br />

1-5<br />

1-5 (hepatic failure)<br />

1-5<br />

>5<br />

>5<br />

57<br />

Lipid Complex<br />

(Abelcet)<br />

3<br />

5<br />

8<br />

5<br />

4<br />

9<br />

8<br />

6<br />

7<br />

b<br />

b<br />

18<br />

14<br />

6<br />

b<br />

b<br />

b<br />

b<br />

4<br />

b<br />

>5 4<br />

1-5<br />

1-5<br />

11<br />

5<br />

Liposomal<br />

(AmBisome)<br />

12<br />

14<br />

14.3<br />

14.3<br />

7.9<br />

13.4<br />

19.8<br />

39.7<br />

31.8<br />

30.3<br />

47.5<br />

19.8<br />

17.2<br />

ALT 14.6<br />

AST 12.8<br />

24.8<br />

10.8<br />

22.4 (creatinine<br />

increase)<br />

>5<br />

14<br />

bAdverse event reported; specific percentages not available<br />

*Only most common adverse events are reported here.<br />

**Rash tends to appear more frequently in immunocompromised patients receiving immunosuppressive medications.<br />

1-5<br />

>5<br />

>5<br />

>5<br />

b<br />

b<br />

b<br />

b<br />

11<br />

7<br />

Fluconazole<br />

Itraconazole<br />

oral capsules<br />

(Systemic,<br />

onychomycosis)<br />

b 0, 3<br />

1.7<br />

3.7<br />

1.7<br />

1.5<br />

1.0<br />

1.9<br />

b<br />

b<br />

b<br />

4<br />

1<br />

3, 2<br />

2, 4 (general GI)<br />

11<br />

5<br />

3<br />

1<br />

2<br />

3<br />

3<br />

4, 1<br />

1, 1<br />

0, 1<br />

b<br />

b<br />

3, 4<br />

b<br />

1.8 9**, 3<br />

3, 3<br />

b<br />

b<br />

1 (albuminuria)<br />

1


Table 10. Common Adverse Events (%), by System, Reported for the Antifungal Agents 23<br />

Adverse Event Ketoconazole Caspofungin Flucytosine Nystatin Terbinafine Voriconazole Griseofulvin<br />

Body as a Whole<br />

Chest Pain<br />

Generalized Pain<br />

Hypertriglyceridemia<br />

Cardiovascular<br />

Edema<br />

Hemorrhage<br />

Hypotension<br />

Hypertension<br />

Tachycardia<br />

Digestive System<br />

Abdominal Pain<br />

Nausea<br />

Vomiting<br />

Diarrhea<br />

Dyspepsia<br />

Appetite decrease<br />

Taste disturbance<br />

Flatulence<br />

Cramping Epigastric Pain<br />

Central Nervous System<br />

Chills/shaking<br />

Dizziness<br />

Fatigue<br />

Fever<br />

Headache<br />

Hallucinations<br />

Malaise<br />

Vertigo<br />

Insomnia<br />

Emotional Lability<br />

Nervousness<br />

Depression<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Cholestatic jaundice<br />

Hepatitis<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Hematologic<br />

Neutropenia<br />

Anemia<br />

Thrombocytopenia<br />

Renal<br />

Abnormal kidney function<br />

Acute kidney failure<br />

Other<br />

Allergic reaction<br />

Decreased libido<br />

Visual disturbance<br />

Weight Loss<br />

Injection Site Pain/Rxn<br />

Face Edema<br />

Weight Gain<br />

Multiple Organ Failure<br />

Sepsis<br />

Rare<br />

1.2<br />

3<br />

3<br />


23- 25<br />

Table 11. Dosing for the Antifungal Agents<br />

Availability<br />

Dose /Frequency/Duration<br />

Amphotericin B<br />

Deoxycholate (Fungizone ® )<br />

50mg vials Infection Treatment regimen Dose 1<br />

(mg/kg/day)<br />

Aspergillosis up to 3.6 g total dose 1 to 1.5<br />

Blastomycosis 4 to 12 weeks 0.5 to 0.6<br />

Candidiasis 4 to 12 weeks 0.5 to 1<br />

Coccidioidomycosis 4 to 12 weeks 0.5 to 1<br />

Cryptococcosis 4 to 12 weeks 0.5 to 0.7<br />

Histoplasmosis 4 to 12 weeks 0.5 to 0.6<br />

Mucormycosis 4 to 12 weeks 1 to 1.5<br />

Rhinocerebral<br />

phycomycosis<br />

3 to 4 g total dose 0.25 to 0.3 up to 1 to 1.5<br />

Sporotrichosis up to 2.5 g total dose 0.5<br />

Amphotericin B Cholesteryl<br />

Sulfate Complex<br />

(Amphotec ® )<br />

Amphotericin B Lipid<br />

Complex (Abelcet ® )<br />

Amphotericin B Liposomal<br />

(AmBisome ® )<br />

Caspofungin Acetate<br />

(Cancidas ®)<br />

Powder for injection:<br />

50mg in 20ml vial and<br />

100mg in 50ml vial,<br />

single use only<br />

Suspension for injection:<br />

100mg/20ml, in 10 and<br />

20ml single use vial<br />

Powder for injection:<br />

50mg, single use vial<br />

Powder for injection:<br />

70mg and 50mg single<br />

use vials<br />

A test dose may be preferred due to patient variability in tolerance: 1mg in<br />

20 ml of 5% dextrose delivered IV over 20 to 30 minutes.<br />

1<br />

Some dosages not FDA Approved.<br />

Fungal infection, systemic:<br />

A test dose is advisable (e.g., 10 ml of final preparation containing 1.6 to<br />

8.3 mg infused over 15 to 30 min). The recommended dose is 3 to<br />

4mg/kg/day prepared as a 0.6mg/ml (range, 0.16 to 0.83mg/ml) infusion<br />

delivered at a rate of 1mg/kg/hr. Do not filter or use an in-line filter.<br />

Fungal infection, systemic:<br />

The recommended dose is 5mg/kg/day prepared as a 1mg/ml infusion<br />

and delivered at a rate of 2.5mg/kg/hour. For pediatric patients and<br />

patients with cardiovascular disease, the drug may be diluted to a final<br />

concentration of 2mg/ml. If the infusion exceeds 2 hours, mix the<br />

contents by shaking the infusion bag every 2 hours. Do not use an in-line<br />

filter.<br />

Fungal infection, empirical:<br />

3mg/kg/day of liposomal amphotericin B using a controlled infusion<br />

device over 120 minutes; infusion time may be reduced to 60 minutes if<br />

well tolerated.<br />

Fungal infection, systemic:<br />

3 to 5mg/kg/day prepared as a 1 to 2mg/ml infusion delivered initially<br />

over 120 minutes; infusion time may be reduced to 60 minutes if well<br />

tolerated. Lower infusion concentrations of 0.2 to 0.5 mg/ml may be<br />

appropriate for infants and small children to provide sufficient volume<br />

for infusion.<br />

Cryptococcal meningitis in HIV:<br />

6mg/kg/day using a controlled infusion device over 120 minutes;<br />

infusion time may be reduced to 60 minutes if well tolerated or increased<br />

if patient experiences discomfort.<br />

Leishmaniasis:<br />

3mg/kg/day on days 1 through 5, 14, and 21 to immunocompetent<br />

patients; a repeat course of therapy may be useful if parasitic clearance is<br />

not achieved. Administer 4mg/kg/day on days 1 through 5, 10, 17, 24,<br />

31, and 38 to immunosuppressed patients; seek expert advice regarding<br />

further therapy if parasitic clearance is not achieved.<br />

Loading dose: 70mg should be administered on day 1<br />

Maintenance: 50 mg daily<br />

Administer by slow IV infusion of 1 hour. Duration of treatment should be<br />

based upon the severity of the patient's underlying disease, recovery from<br />

immunosuppression, and clinical response. The efficacy of a 70 mg dose<br />

regimen in patients who are not clinically responding to the 50 mg daily<br />

dose is not known. Limited safety data suggests that an increase in dose to<br />

59


Fluconazole (Diflucan ® )<br />

Injection: 2mg/ml in<br />

100ml and 200ml<br />

Oral: 50mg, 100mg,<br />

150mg, 200mg tablets;<br />

powder for oral<br />

suspension in 10mg/ml<br />

and 40mg/ml<br />

70 mg daily is well tolerated. The safety and efficacy of doses >70 mg have<br />

not been adequately studied.<br />

Dosing for oral and IV administration is the same.<br />

Adults:<br />

Single-dose:<br />

Vaginal candidiasis:<br />

150mg as a single oral dose.<br />

Oropharyngeal candidiasis:<br />

200mg on the first day, followed by 100mg once daily for 2 weeks<br />

to decrease the likelihood of relapse.<br />

Esophageal candidiasis:<br />

200mg on the first day, followed by 100mg once daily. Doses up<br />

to 400 mg/day may be used based on response. Duration should<br />

be a minimum of 3 weeks and for 2 weeks or more following<br />

resolution of symptoms.<br />

Candidiasis, other:<br />

For candidal urinary tract infections (UTIs) and peritonitis, 50 to<br />

200mg/day has been used. For systemic candidal infections<br />

(including candidemia, disseminated candidiasis, and pneumonia),<br />

optimal dosage and duration have not been determined, although<br />

doses of 400mg/day or less have been used.<br />

Prevention of candidiasis in bone marrow transplant:<br />

400mg once daily. In patients anticipated to have severe<br />

granulocytopenia (less than 500neutrophils/mm 3 ), start<br />

fluconazole prophylaxis several days before anticipated onset of<br />

neutropenia, and continue 7 days after neutrophil count rises<br />

above 1000cells/mm 3 .<br />

Cryptococcal meningitis:<br />

400mg on the first day followed by 200mg once daily. A dosage<br />

of 400mg once daily may be used based on the patient's response<br />

to therapy. The duration of treatment for initial therapy of<br />

cryptococcal meningitis is 10 to 12 weeks after the cerebrospinal<br />

fluid (CSF) becomes culture negative. The dosage of fluconazole<br />

for suppression of relapse of cryptococcal meningitis in patients<br />

with AIDS is 200mg once daily.<br />

60<br />

Children:<br />

Equivalent Fluconazole Dosage in<br />

Children vs. Adults<br />

Children Adults<br />

3 mg/kg 100 mg<br />

6 mg/kg 200 mg<br />

12 mg/kg 1 400 mg<br />

Neonates:<br />

Experience in neonates is limited to pharmacokinetic studies in<br />

premature newborns. Based on the prolonged half-life seen in<br />

premature newborns (gestational age, 26 to 29 weeks), administer<br />

the same dosage (mg/kg) to neonates in the first 2 weeks of life as<br />

in older children, but administer every 72 hours. After the first 2<br />

weeks, dose neonates once daily.<br />

Oropharyngeal candidiasis:<br />

6mg/kg on the first day, followed by 3mg/kg once daily.<br />

Administer treatment for at least 2 weeks to decrease the<br />

likelihood of relapse.<br />

Esophageal candidiasis:<br />

6mg/kg on the first day followed by 3mg/kg once daily. Doses up<br />

to 12mg/kg/day may be used based on the patient's response to<br />

therapy. Treat patients with esophageal candidiasis for a<br />

minimum of 3 weeks and for at least 2 weeks following the<br />

resolution of symptoms.<br />

Systemic Candida infections:<br />

Daily doses of 6 to 12mg/kg/day have been used in an open,


61<br />

noncomparative study of a small number of children.<br />

Cryptococcal meningitis:<br />

12mg/kg on the first day, followed by 6mg/kg once daily. A<br />

dosage of 12mg/kg once daily may be used based on medical<br />

judgment of the patient's response to therapy. The recommended<br />

duration of treatment for initial therapy of cryptococcal meningitis<br />

is 10 to 12 weeks after the CSF becomes culture negative. For<br />

suppression of cryptococcal meningitis relapse in children with<br />

AIDS, the recommended dose is 6 mg/kg once daily.<br />

1 Some older children may have clearances similar to that of adults. Absolute doses<br />

more than 600mg/day are not recommended.<br />

Flucytosine (Ancobon ® ) 250 and 500mg capsules Usual dosage is 50 to 150mg/kg/day in divided doses at 6-hour intervals. To<br />

reduce or avoid nausea or vomiting, take capsules a few at a time over a 15-<br />

minute period.<br />

Itraconazole (Sporanox ® )<br />

Injection: 10mg/ml (kit)<br />

Oral: 100mg capsules,<br />

10mg/ml oral<br />

solution<br />

Itraconazole oral solution and capsules cannot be used interchangeably.<br />

Itraconazole capsules should be taken with a full meal to ensure absorption.<br />

The oral solution should be taken without food if possible.<br />

Capsules:<br />

Aspergillosis: A daily dose of 200 to 400mg is recommended.<br />

Blastomycosis/histoplasmosis:<br />

200mg once daily. If there is no obvious improvement or there is<br />

evidence of progressive fungal disease, increase the dose in<br />

100mg increments to a maximum of 400mg/day. Give doses<br />

above 200mg/day in 2 divided doses.<br />

Life-threatening situations:<br />

Although clinical studies did not provide for a loading dose, it is<br />

recommended, based on pharmacokinetic data, that a loading dose<br />

of 200mg 3 times/day be given for the first 3 days of treatment.<br />

Continue treatment for a minimum of 3 months (at 200-400mg<br />

QD-BID) and until clinical parameters and laboratory tests<br />

indicate that the active fungal infection has subsided. An<br />

inadequate period of treatment may lead to recurrence of active<br />

infection.<br />

Treatment of onychomycosis (fingernails only):<br />

Two treatment pulses, each consisting of 200mg twice daily for 1<br />

week. The pulses are separated by a 3-week period without<br />

itraconazole.<br />

Treatment of onychomycosis (toenails with or without fingernail<br />

involvement): 200mg/day for 12 weeks.<br />

Oral Solution:<br />

Esophageal candidiasis:<br />

100mg/day for a minimum treatment of 3 weeks. Continue<br />

treatment for 2 weeks following resolution of symptoms. Doses up<br />

to 200mg/day may be used based on medical judgment of the<br />

patient's response to therapy. Vigorously swish the solution in the<br />

mouth (10mL at a time) for several seconds and swallow.<br />

Empiric therapy in neutropenic patients with suspected fungal<br />

infections:<br />

After approximately 14 days of IV therapy, continue treatment<br />

with oral solution 200mg twice daily until resolution of clinically<br />

significant neutropenia. The safety and efficacy of itraconazole<br />

use exceeding 28 days in this population is not known.<br />

Oropharyngeal candidiasis:<br />

200mg/day for 1 to 2 weeks. Vigorously swish the solution in the<br />

mouth (10ml at a time) for several seconds and swallow. For<br />

patients with oropharyngeal candidiasis unresponsive/refractory to<br />

treatment with fluconazole tablets, the recommended dose of<br />

itraconazole is 100mg twice daily. Expect clinical response in 2 to<br />

4 weeks. Patients may be expected to relapse shortly after<br />

discontinuing therapy. Limited data on the safety of long-term use<br />

(more than 6 months) of the oral solution are available at this time.


Injection:<br />

Use only the components provided in the kit for preparation of the injection.<br />

Blastomycosis, histoplasmosis, and aspergillosis:<br />

200mg IV twice daily for 4 doses, followed by 200mg/day. Infuse<br />

each IV dose over 1 hour. Itraconazole can be given as oral<br />

capsules or IV. The safety and efficacy of the injection<br />

administered for more than 14 days are not known.<br />

Continue total itraconazole therapy (injection followed by<br />

capsules) for a minimum of 3 months and until clinical parameters<br />

and laboratory tests indicate that the active fungal infection has<br />

subsided. An inadequate period of treatment may lead to<br />

recurrence of active infection.<br />

Empiric therapy in febrile neutropenic patients with suspected fungal<br />

infections:<br />

200mg twice daily for 4 doses, followed by 200mg once daily for<br />

up to 14 days. Infuse each IV dose over 1 hour. Continue<br />

treatment with 200mg itraconazole oral solution (20 ml) twice<br />

daily until resolution of clinically significant neutropenia. The<br />

safety and efficacy of itraconazole use exceeding 28 days in ETFN<br />

is not known.<br />

Ketoconazole (Nizoral ® ) 200mg tablets Adults:<br />

Initially, 200mg once daily. In very serious infections, or if clinical<br />

response is insufficient, increase dose to 400mg once daily.<br />

Children:<br />

(> 2 years of age):<br />

3.3 to 6.6mg/kg/day as a single daily dose.<br />

(< 2 years of age):<br />

Daily dosage has not been established.<br />

Minimum treatment for candidiasis is 1 or 2 weeks and 6 months for the<br />

other indicated systemic mycoses. Chronic mucocutaneous candidiasis<br />

usually requires maintenance therapy. Minimum treatment of recalcitrant<br />

dermatophyte infections is 4 weeks in cases involving glabrous skin. Palmar<br />

and plantar infections may respond more slowly. Apparent cures may<br />

subsequently recur after discontinuation of therapy in some cases.<br />

Nystatin (Mycostatin ® )<br />

500,000unit tablets<br />

100,000units/ml oral<br />

suspension<br />

200,000unit<br />

pastilles/troches<br />

Oral powder for<br />

suspension:<br />

50 million units<br />

150 million units<br />

500 million units<br />

1 billion units<br />

2 billion units<br />

5 billion units<br />

Oral tablets for nonesophageal GI candidiasis: 1 to 2 tablets (500,000<br />

to 1,000,000units) TID continued for 48 hours after clinical cure to prevent<br />

relapse.<br />

Oral Suspension, adults and children: 400,000 to 600,000units QID<br />

(one half of dose in each side of mouth, retaining the drug as long as<br />

possible before swallowing). Infants: 200,000units 4 times daily<br />

(100,000units in each side of mouth).<br />

Pastilles / troches: 200,000 to 400,000units 4-5 times daily for up to<br />

14 days or until 48 hours after disappearance of oral symptoms.<br />

This dosage form must be allowed to dissolve slowly in the mouth,<br />

not chewed or swallowed whole<br />

Powder for extemporaneous compounding:<br />

Adults and children:<br />

Add 1/8 tsp ( 500,000units) to 1/2 cup of water and stir well.<br />

Administer 4 times daily. Use immediately after mixing; do not store.<br />

Continue local treatment at least 48 hours after perioral signs and<br />

symptoms have disappeared and cultures have returned to normal.<br />

To improve oral retention of the drug, nystatin (250,000units) has been<br />

administered for oral candidiasis in the form of flavored frozen popsicles.<br />

Terbinafine (Lamisil ® ) 250mg tablet Onychomycosis:<br />

Fingernail= 250mg QD for 6 weeks<br />

Toenail= 250mg QD for 12 weeks<br />

Voriconazole (Vfend ® )<br />

Oral: 50mg and 200mg<br />

tablets; powder for oral<br />

62<br />

Invasive aspergillosis and serious fungal infections:<br />

Loading dose= 6 mg/kg IV every 12 hours for 2 doses


suspension 40mg/ml<br />

Powder for injection:<br />

200mg, single use vials<br />

Maintenance dose= 4 mg/kg IV every 12 hours<br />

Once the patient can tolerate oral medication, the tablet form or<br />

oral suspension of voriconazole may be used at a dose of 200mg<br />

every 12 hours in patients who weigh more than 40kg or in those<br />

who weigh less than 40kg, the maintenance dose is 100mg every<br />

12 hours. If patient response is inadequate, the oral maintenance<br />

dose may be increased from 200 mg every 12 hours to 300mg<br />

every 12 hours and for patients weighing less than 40kg, the oral<br />

maintenance dose may be increased from 100mg every 12 hours<br />

to 150mg every 12 hours.<br />

Griseofulvin Microsize<br />

(Fulvicin ® U/F,<br />

Grifulvin ® V)<br />

Griseofulvin Ultramicrosize<br />

(Gris-PEG ® ,<br />

Fulvicin ® PG)<br />

250mg capsule<br />

250mg and 500mg tablet<br />

125mg and 250mg tablet<br />

and oral suspension<br />

125mg/5ml with alcohol<br />

0.2% (120ml)<br />

125 and 250mg tablet<br />

165mg, 250mg and<br />

330mg tablet<br />

Esophageal Candidiasis:<br />

Oral dose of 200mg every 12 hours for patients who weigh 40kg<br />

or more. Adult patients who weigh less than 40kg should receive<br />

an oral dose of 100mg every 12hours. Treatment duration is a<br />

minimum of 14 days and for at least seven days following<br />

resolution of symptoms.<br />

Note: Voriconazole tablets should be taken at least one hour<br />

before or one hour following a meal. Switching between IV and<br />

oral is appropriate when necessary.<br />

Children:<br />

Microsize: 10-20mg/kg/day in single or 2 divided doses<br />

Ultramicrosize: >2 years: 5-10mg/kg/day in single or 2 divided<br />

doses<br />

Adults:<br />

Microsize: 500-1000mg/day in single or divided doses<br />

Ultramicrosize: 330-375mg/day in single or divided doses; doses up<br />

to 750mg/day have been used for infections more difficult to<br />

eradicate such as tinea unguium<br />

Duration of therapy:<br />

Tinea corporis: 2-4 weeks<br />

Tinea capitis: 4-6 weeks or longer<br />

Tinea pedis: 4-8 weeks<br />

Tinea unguium: 3-6 months or longer<br />

Note: The efficiency of GI absorption of griseofulvin ultramicrosize is 1.5 times<br />

that of conventional microsized griseofulvin. This factor permits the oral intake of 2/3<br />

as much ultramicrosize as the microsize form, but there is no evidence that this<br />

confers any significant clinical difference in regard to safety and efficacy.<br />

63


Special Dosing Considerations<br />

In studies where patients experienced nephrotoxicity while receiving conventional amphotericin<br />

B, and a lipid formulation of amphotericin B was substituted, serum creatinine concentrations<br />

23, 24<br />

generally declined during therapy with the lipid-based or liposomal formulations.<br />

Table 12. Special Dosing Considerations for the Antifungal Agents 23-25<br />

Drug Renal Dosing Hepatic<br />

Dosing<br />

Pediatric Use<br />

Amphotericin B<br />

No<br />

Deoxycholate<br />

Amphotericin B<br />

Cholesteryl<br />

Sulfate Complex<br />

Amphotericin B<br />

Lipid Complex<br />

Amphotericin B<br />

Liposomal<br />

Caspofungin<br />

Acetate<br />

Yes (Alternate<br />

daily dosing at<br />

1.5mg/kg/day);<br />

hydration and<br />

sodium repletion<br />

may decrease the<br />

risk of<br />

nephrotoxicity<br />

Not established by<br />

adequate wellcontrolled<br />

trials.<br />

No No No unusual adverse<br />

events have been<br />

reported with use in<br />

children (50ml/min =<br />

100% of usual<br />

daily dose and<br />


Itraconazole<br />

40ml/min., Q24<br />

hours with Cr. Cl.<br />

of 10-20ml/min,<br />

and Q24-48 hours<br />

or longer with Cr.<br />

Cl


Terbinafine<br />

Voriconazole<br />

Griseofulvin<br />

Ultramicrosize<br />

Griseofulvin<br />

Microsize<br />

Clearance of terbinafine may be<br />

decreased (by 50%) by patients with<br />

renal impairment or in those with<br />

preexisting liver disease.<br />

Terbinafine has not been adequately<br />

studied in this population, therefore,<br />

the manufacturer does not<br />

recommend use in either population.<br />

In moderatesevere<br />

renal<br />

failure, IV<br />

voriconazole<br />

should be used<br />

only when clearly<br />

needed due to<br />

accumulation of<br />

the IV vehicle; no<br />

adjustment is<br />

needed for the<br />

oral formulation<br />

In mildmoderate<br />

hepatic<br />

disease, the<br />

usual IV or<br />

oral loading<br />

dose should be<br />

used,<br />

however, the<br />

IV and oral<br />

maintenance<br />

doses should<br />

be decreased<br />

by 50%<br />

No dose adjustments are discussed<br />

in the manufacturers labeling,<br />

however, griseofulvin is<br />

contraindicated in patients with<br />

porphyria and hepatocellular failure.<br />

The safety and<br />

efficacy of<br />

terbinafine has not<br />

been established in<br />

pediatric patients<br />

(


VIII. Comparative Effectiveness of the Antifungal Agents<br />

The available lipid formulations of amphotericin B represent advancement in drug delivery technology. Clinical experience has primarily been in patients either<br />

refractory to or intolerance of conventional amphotericin B deoxycholate. 28 The lipid-based products have not demonstrated superior efficacy when<br />

prospectively compared with traditional amphotericin B in the treatment of documented infections. In some patients with life-threatening mycosis who failed<br />

treatment with, or were intolerant to, amphotericin B, the lipid formulations have been effective. 27<br />

When used for empirical treatment of febrile neutropenia, amphotericin B liposomal (AmBisome) significantly reduced the incidence of proven emergent fungal<br />

infections but did not improve short-term survival rates, in comparison with traditional amphotericin B. 34 For these reasons, the lipid amphotericin B products<br />

can be restricted for use in those who are intolerant of or refractory to amphotericin B.<br />

Table 13. Additional Outcomes Evidence for the Antifungal Agents<br />

Study Design Entry Criteria Sample Treatment<br />

Regimen<br />

Miller CB, et<br />

al. 35<br />

Fleming RV,<br />

et al. 36<br />

Retrospective<br />

analysis<br />

Randomized,<br />

double blind<br />

study<br />

• Adult hematopoietic<br />

stem cell transplant<br />

recipients<br />

• Proven or probable<br />

invasive fungal<br />

infection<br />

• Transplantation<br />

between 1995-2000<br />

Patients with leukemia<br />

and suspected or<br />

documented mycosis<br />

- Amphotericin B<br />

deoxycholate vs.<br />

AmBisome<br />

vs.<br />

Abelcet<br />

n=75 ABLC or<br />

AmBisome at 3-<br />

5mg/kg/day<br />

Duration of<br />

Study<br />

Amphotericin B Studies<br />

Infection and<br />

response<br />

dependent<br />

10 days at 3<br />

mg/kg for<br />

ABLC and 15<br />

days at 4 mg/kg<br />

for AmBisome<br />

Results<br />

In assessing renal function based on medical records in adult hematopoietic stem<br />

cell transplant recipients with proven or probable invasive fungal infection:<br />

• Due to nephrotoxicity, 88% of patients treated with amphotericin B<br />

deoxycholate were switched to a lipid formulation of amphotericin B.<br />

• 53% of patients initiated on amphotericin B were switched within the<br />

first week of therapy.<br />

• Significantly more patients (71%) treated with amphotericin B<br />

experienced a 100% increase in serum creatinine from baseline<br />

compared to patients treated with either AmBisome (44%) or Abelcet<br />

(41.2%).<br />

• Statistical analysis showed that compared to patients initiated on<br />

AmBisome or Abelcet, the risk of nephrotoxicity, dialysis, and death<br />

were all increased for patients initiated on amphotericin B<br />

deoxycholate.<br />

In evaluating the efficacy of ABLC and AmBisome:<br />

• The overall response to therapy was 27/43 (63%) for ABLC and 15/39<br />

(39%) for AmBisome (p=0.03).<br />

• Response rates were approximately 30% in both groups for patients<br />

with documented fungal infections.<br />

• Acute, not dose-limiting infusion side effects were seen in 70% vs.<br />

36% (p=0.002), ABLC vs. AmBisome.<br />

• Severe initial infusion reactions, all leading to discontinuation,<br />

occurred in three cases (one with ABLC and two with AmBisome).<br />

• Increase of bilirubin > 1.5 times from baseline was 38% vs. 59%,<br />

67


Cagnoni PJ 37<br />

Subira SM, et<br />

al. 38<br />

Kontoyiannis<br />

DP, et al. 39<br />

Randomized,<br />

double blind,<br />

multicenter<br />

trial<br />

Randomized,<br />

controlled<br />

trial<br />

Meta-analysis<br />

of prospective<br />

studies<br />

• Patients with febrile<br />

neutropenia<br />

• Empiric antifungal<br />

therapy<br />

• Patients with<br />

hematologic<br />

malignancies and<br />

fever of unknown<br />

origin after<br />

chemotherapy or<br />

autologous stem cell<br />

transplantation<br />

• Empiric therapy<br />

n=414 Liposomal<br />

amphotericin B<br />

vs. conventional<br />

amphotericin B<br />

n=105 ABLC at<br />

1mg/kd/day or<br />

conventional<br />

amphotericin B at<br />

0.6mg/kg/day<br />

Adults with candidaemia - Fluconazole vs.<br />

amphotericin B<br />

Infection and<br />

response<br />

dependent<br />

Infection and<br />

response<br />

dependent<br />

ABLC vs. AmBisome (p=0.05).<br />

• ABLC and AmBisome were equally effective for the treatment of<br />

suspected or documented fungal infections.<br />

• While, acute infusion-toxicity was greater with ABLC, infusion<br />

toxicity requiring discontinuation was similar for both drugs.<br />

• AmBisome was better tolerated than ABLC but was associated with<br />

mild abnormalities in liver function tests at the end of therapy.<br />

Using a composite end-point, liposomal amphotericin B and conventional<br />

amphotericin B were found to be equivalent in overall efficacy. Other findings<br />

included:<br />

• The liposomal amphotericin B treatment group had fewer proven<br />

fungal infections, fewer infusion-related side effects, and less<br />

nephrotoxicity.<br />

• Authors commented on findings of a randomized, double-blind study<br />

comparing liposomal amphotericin B at 3 or 5 mg/kg/day with<br />

amphotericin B lipid complex (ABLC) at 5mg/kg/day as empiric<br />

therapy in patients with febrile neutropenia. Liposomal amphotericin B<br />

was associated with less toxicity than ABLC, both in terms of infusionrelated<br />

reactions and nephrotoxicity.<br />

• In the previously mentioned study, the incidence of study drug<br />

discontinuation due to toxicity was: liposomal amphotericin B<br />

3mg/kg/day 14%; liposomal amphotericin B 5mg/kg/day 15%; and<br />

ABLC 42% (p


Johansen HK,<br />

et al. 40<br />

Meta-analysis<br />

of 16 trials,<br />

looking at the<br />

effect of<br />

morbidity and<br />

mortality<br />

Patients with cancer,<br />

complicated with<br />

neutropenia<br />

n=3,760<br />

Fluconazole vs.<br />

amphotericin B<br />

and microbiological failure according to all Candida species (OR, 0.99;<br />

CI, 0.78-1.26).<br />

• A trend favoring amphotericin B was seen in mycological eradication<br />

of non-albicans Candida species (OR, 0.70; CI, 0.47-1.06).<br />

• Amphotericin B was more toxic than fluconazole (OR, 2.94; CI, 2.14-<br />

4.4).<br />

• Summary: fluconazole is as efficacious as and less toxic than<br />

amphotericin B in stable, not severely immunosuppressed candidaemic<br />

patients at low risk for death. However, fluconazole may be less<br />

effective than amphotericin B in candidaemias caused by some nonalbicans<br />

Candida species.<br />

- Sixteen trials were included for comparative analysis:<br />

• In three large 3-armed trials, results for amphotericin B were combined<br />

with results for nystatin in a "polyene" group. Because nystatin is an<br />

ineffective drug in these circumstances, this approach creates a bias in<br />

favor of fluconazole. Furthermore, most patients were randomized to<br />

oral amphotericin B, which is poorly absorbed and poorly documented.<br />

It was unclear whether there was overlap among the "polyene" trials.<br />

• There were no significant differences in effect between fluconazole and<br />

amphotericin B, but the confidence intervals were wide.<br />

• More patients dropped out of the study when they received<br />

amphotericin B, but as none of the trials were blinded, decisions on<br />

premature interruption of therapy could have been biased.<br />

• Furthermore, amphotericin B was rarely given under optimal<br />

circumstances, with premedication to reduce infusion-related toxicity,<br />

slow infusion, and with potassium and magnesium supplements to<br />

prevent nephrotoxicity.<br />

• Summary: Amphotericin B has been disfavored in several trials due to<br />

design or analysis. Since intravenous amphotericin B is the only<br />

antifungal agent for which there is good evidence suggesting an effect<br />

on mortality, consideration should be given for its use.<br />

Bowden R, et<br />

al. 41<br />

Randomized,<br />

double-blind,<br />

multicenter<br />

trial<br />

Invasive aspergillosis n=174 Amphotec<br />

6mg/kg/day vs.<br />

conventional<br />

amphotericin B 1-<br />

1.5mg/kg/day<br />

Infection and<br />

response<br />

dependent<br />

In comparing the effects of amphotericin B colloidal dispersion (Amphotec,<br />

ABCD) to conventional amphotericin B (AmB) in the treatment of invasive<br />

aspergillosis:<br />

• For evaluable patients in the ABCD and AmB treatment groups,<br />

respective rates of therapeutic response (52% vs. 51%; p=1.0),<br />

mortality (36% vs. 45%; p=.4), and death due to fungal infection (32%<br />

vs. 26%; p=.7) were similar.<br />

• Renal toxicity was lower (25% vs. 49%; p=.002) and the median time<br />

69


Barrett JP, et<br />

al. 42 Meta-analysis Literature search of<br />

Medline, Embase,<br />

Biological Abstracts,<br />

Aidsline, Cancerlit, CRD<br />

database, Cochrane<br />

Controlled Trials<br />

Register, and other<br />

databases.<br />

Wingard JR,<br />

et al. 43<br />

Clark AD, et<br />

al. 44<br />

Randomized,<br />

double-blind<br />

comparative<br />

trial<br />

Retrospective<br />

analysis<br />

Neutropenic patients<br />

with unresolved fever<br />

after 3 days of<br />

antibacterial therapy<br />

• Single-center, 5-<br />

year use of<br />

amphotericin B<br />

8 publications<br />

n=244 ABLC<br />

5mg/kg/day vs.<br />

liposomal<br />

amphotericin B<br />

(L Amph) at 3<br />

and 5mg/kg/day<br />

- AmBisome vs.<br />

Abelcet<br />

to onset of nephrotoxicity was longer (301 vs. 22 days; p


Linder N, et<br />

al. 45<br />

Kartsonis<br />

NA, et al. 46<br />

Cesaro S, et<br />

al. 47<br />

Colombo<br />

AL, et al. 48<br />

Comparative<br />

study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Randomized,<br />

double blind,<br />

• Focus on<br />

indications,<br />

efficacy, and<br />

toxicity<br />

Infants in the neonatal<br />

intensive care unit from<br />

1996-2000 with candida<br />

bloodstream infections<br />

• Patients with<br />

mucosal or invasive<br />

candida infections<br />

• Refractory or<br />

intolerant to<br />

amphotericin B or<br />

lipid amphotericin B<br />

• Invasive mycosis, as<br />

maintenance or<br />

rescue therapy, for<br />

refractory infection<br />

not responsive to<br />

liposomal<br />

amphotericin B or<br />

fluconazole<br />

• Diagnosis of<br />

patients included:<br />

leukemia,<br />

myelodysplastic<br />

syndrome, and<br />

Hodgkin’s<br />

n=56 Conventional<br />

amphotericin B,<br />

liposomal<br />

amphotericin B<br />

(LamB), and<br />

amphotericin B<br />

colloidal<br />

dispersion<br />

(ABCD)<br />

n=37 Caspofungin<br />

n=10 Caspofungin and<br />

liposomal<br />

amphotericin B<br />

followed by<br />

voriconazole<br />

lymphoma<br />

Invasive candidiasis n=239 Caspofungin vs.<br />

amphotericin B<br />

If blood cultures<br />

were + more<br />

than 10 days<br />

with clinical<br />

signs of fungal<br />

infection, a<br />

second<br />

antifungal was<br />

initiated.<br />

Caspofungin<br />

Infection and<br />

response<br />

dependent<br />

Combination<br />

therapy with<br />

caspofungin and<br />

liposomal<br />

amphotericin B<br />

was given for a<br />

median of 17<br />

days (range 6-<br />

40).<br />

Voriconazole<br />

was given for a<br />

mean of 75 days<br />

(range 42-194)<br />

4.8mg/kg/day.<br />

• Nephrotoxicity and electrolyte abnormalities were similar in both<br />

groups.<br />

• Rigors and febrile episodes were more common with Abelcet.<br />

In comparing conventional amphotericin B, liposomal amphotericin B, and<br />

amphotericin B colloidal dispersion:<br />

• No differences in mortality were found between the three groups.<br />

• Sterilization of the blood was achieved with amphotericin B in 67.6%<br />

of patients, LamB in 83.3%, and ABCD in 57.1%, when used as<br />

monotherapy; with the addition of a second antifungal agent, success<br />

rates were 100%, 83.3%, and 92.8%, respectively.<br />

• There were no differences between the groups in the time to resolution<br />

of fungaemia.<br />

• No patients had immediate local or systemic adverse events and none<br />

showed deterioration in renal function.<br />

In evaluating caspofungin in patients who are refractory to treatment with >/= 1<br />

antifungal agent:<br />

• A favorable response was noted in 82% (14/17) of patients with<br />

esophageal candidiasis, 100% (4/4) with oropharyngeal candidiasis, and<br />

87% (13/15) with invasive candidiasis.<br />

• Caspofungin was generally well tolerated; one serious drug-related<br />

adverse event was reported.<br />

In evaluating the safety and efficacy of caspofungin and liposomal amphotericin<br />

B, followed by voriconazole:<br />

• Among the nine patients with proven or probable mycosis, one was not<br />

evaluated because of early death caused by massive hemoptysis whilst<br />

in the remaining eight patients, the response was classified as complete,<br />

stable and failure in four, three, and one patients, respectively.<br />

• Complete response was also observed in patients with possible<br />

mycosis.<br />

• Voriconazole was well tolerated although some drug interactions were<br />

observed during treatment with methotrexate and digoxin.<br />

• Overall, a favorable response to antifungal treatment (including the<br />

case of possible mycosis) was obtained in 8 of 10 patients.<br />

In comparing caspofungin with amphotericin B for invasive candidiasis:<br />

• In the USA and Canada, Candida glabrata was the second most<br />

71


international<br />

study<br />

Mora-Duarte Double blind,<br />

J, et al. 49 randomized<br />

trial<br />

Galgiani JN,<br />

et al. 50<br />

Randomized,<br />

double blind,<br />

placebo<br />

controlled,<br />

multicenter<br />

trial<br />

Primary treatment of<br />

invasive candidiasis,<br />

with a subgroup of<br />

patients with candidemia<br />

Soft tissue, or skeletal<br />

coccidioidal infections<br />

n=239 Caspofungin vs.<br />

amphotericin B<br />

Infection and<br />

response<br />

dependent<br />

Infection and<br />

response<br />

dependent<br />

Oral Fluconazole, Itraconazole, Terbinafine and Ketoconazole<br />

n=198 Fluconazole<br />

400mg QD,<br />

itraconazole<br />

200mg BID, or<br />

Placebo<br />

commonly isolated pathogen (18%). In contrast, Candida parapsilosis<br />

and Candida tropicalis accounted for 55% of cases in Latin America.<br />

• Outcomes were comparable for patients treated with caspofungin (74%<br />

overall; 64% and 80% for infections due to Candida albicans and nonalbicans<br />

species) and amphotericin B (62% overall; 58% and 68% for<br />

infections due to Candida albicans and non- albicans species), and<br />

were generally similar across continents.<br />

In studying the efficacy of caspofungin and amphotericin B:<br />

• A modified intention-to-treat analysis showed that the efficacy of<br />

caspofungin was similar to that of amphotericin B, with successful<br />

outcomes in 73.4 % of the patients treated with caspofungin and in<br />

61.7% of those treated with amphotericin B (difference after<br />

adjustment for APACHE II score and neutropenic status, 12.7<br />

percentage points; 95.6% confidence interval, -0.7 to 26.0).<br />

• An analysis of patients who met prespecified criteria for evaluation<br />

showed that caspofungin was superior, with a favorable response in<br />

80.7%of patients, as compared with 64.9%of those who received<br />

amphotericin B (difference, 15.4 percentage points; 95.6 percent<br />

confidence interval, 1.1 to 29.7).<br />

• Caspofungin was as effective as amphotericin B in patients who had<br />

candidemia, with a favorable response in 71.7% and 62.8% of patients,<br />

respectively (difference, 10.0 percentage points; 95.0 percent<br />

confidence interval, -4.5 to 24.5).<br />

• There were significantly fewer drug-related adverse events in the<br />

caspofungin group than in the amphotericin B group.<br />

• Summary: Caspofungin is at least as effective as amphotericin B for<br />

the treatment of invasive candidiasis and, more specifically,<br />

candidemia.<br />

Primary Endpoint<br />

• Response to therapy defined as a 50% reduction in baseline<br />

abnormalities during the first eight months of therapy.<br />

Efficacy: Fluconazole = Itraconazole<br />

• 50% of patients (47 of 94) and 63% of patients (61 of 97) responded to<br />

eight months of treatment with fluconazole and itraconazole,<br />

respectively (difference, 13 percentage points [95% CI, -2 to 28<br />

percentage points]; p=0.08).<br />

• Patients with skeletal infections responded twice as frequently to<br />

itraconazole as to fluconazole.<br />

• By month 12, 57% of patients had responded to fluconazole and 72%<br />

72


Evans EGV,<br />

et al. 51<br />

Randomized,<br />

double blind,<br />

parallel group,<br />

Multicenter<br />

• 18-75 years of age<br />

• Clinical and<br />

mycological<br />

diagnosis of<br />

dermatophyte<br />

onychomycosis of<br />

the toenail<br />

n=496 Terbinafine<br />

250mg QD for 12<br />

weeks (T 12<br />

group),<br />

terbinafine<br />

250mg QD for 16<br />

weeks (T 16<br />

group),<br />

itraconazole<br />

400mg QD for<br />

one week Q 4<br />

weeks for 12<br />

weeks (I 3 group),<br />

or<br />

itraconazole<br />

400mg QD for<br />

one week Q 4<br />

weeks for 16<br />

weeks (I 4 group)<br />

had responded to itraconazole (difference, 15 percentage points [CI<br />

,0.003 to 30 percentage points]; p=0.05).<br />

• Neither fluconazole nor itraconazole showed statistically superior<br />

efficacy in nonmeningeal coccidioidomycosis.<br />

• Relapse rates after discontinuation of therapy did not differ<br />

significantly between groups (28% after fluconazole treatment and 18%<br />

after itraconazole treatment).<br />

Safety: Fluconazole = Itraconazole<br />

• Serious adverse events occurred in 8 of 97 fluconazole-treated patients<br />

(8% [CI, 4% to 16%] ) and 6 of 101 itraconazole treated patients (6%<br />

[CI, 2% to 12%] (p>0.2).<br />

• Three adverse events may have been caused by a study drug: elevated<br />

liver enzyme levels (one itraconazole treated patient), hypokalemia<br />

(one itraconazole treated patient), and vomiting (one fluconazole<br />

treated patient).<br />

• Six patients treated with fluconazole and two treated with itraconazole<br />

were withdrawn from the study because of rash, dry skin, and<br />

gastrointestinal symptoms (fluconazole) , or difficulty concentrating<br />

(itraconazole).<br />

72 weeks Primary Endpoint<br />

• Assessment of mycological cure at week 72, defined as negative results<br />

on microscopy and culture of samples from the target toenail.<br />

Efficacy: Terbinafine > Itraconazole<br />

• At week 72 the mycological cure rates were 75.7% (81/107) in the T 12<br />

group and 80.8 (80/99) in the T 16 group compared with 38.3% (41/107)<br />

in the I 3 group and 49.1% (53/108) in the I 4 group.<br />

• All comparisons (T 12 vs. I 3 , T 12 vs. I 4 , T 16 vs. I 3 T 16 vs. I 4 ) showed<br />

higher cure rates in the terbinafine groups (all p


Brautigam Randomized,<br />

M, et al. 52 double blind,<br />

parallel group,<br />

Multicenter<br />

study<br />

Ally R, et<br />

al. 53<br />

Randomized,<br />

double blind,<br />

parallel group,<br />

multicenter<br />

• 18 years of age or<br />

older<br />

• Diagnosis of distal<br />

subungual or<br />

proximal<br />

onychomycosis and<br />

growth of<br />

dermatophytes in a<br />

mycological culture<br />

up to 12 weeks after<br />

the start of<br />

treatment<br />

• 18 to 75 years of<br />

age<br />

• Immunocompromised<br />

• Male or nonpregnant<br />

female<br />

• Diagnosis of<br />

esophagitis based<br />

on clinical<br />

symptoms with or<br />

without<br />

concomitant<br />

oropharyngeal<br />

candidiasis<br />

n=170 Terbinafine<br />

250mg QD<br />

vs.<br />

itraconazole<br />

200mg QD<br />

n=391 Voriconazole<br />

200mg bid +<br />

placebo or<br />

Day 1:<br />

fluconazole<br />

400mg + placebo,<br />

Day 2 onward:<br />

fluconazole<br />

200mg + placebo<br />

52 weeks:<br />

12 weeks of<br />

active drug; 40<br />

weeks of follow<br />

up<br />

recorded in the terbinafine or itraconazole groups.<br />

• Most common adverse events: nausea, headache, upper respiratory tract<br />

infection, chest infection, back pain, flu-like symptoms, bronchitis and<br />

fever.<br />

Primary Endpoints<br />

• Mycological cure (negative results on microscopy and culture) and<br />

clinical improvement (length and area of unaffected nail) at week 52 or<br />

at discontinuation of treatment.<br />

Efficacy: Terbinafine > Itraconazole<br />

• Mycological cure rates were 81% (70 out of 86) for terbinafine and<br />

63% (53 out of 84) for itraconazole (p


Oude Lashof<br />

AM, et al. 54<br />

Gupta AK,<br />

et al. 55<br />

Multicenter,<br />

randomized,<br />

comparative<br />

study<br />

Cumulative<br />

meta-analysis<br />

of 36<br />

(Esophageal<br />

candidiasis was<br />

confirmed by<br />

esophagoscopy,<br />

plus positive<br />

microscopy and<br />

mycological<br />

culture from a<br />

brush biopsy or<br />

tissue biopsy of<br />

esophageal<br />

lesions.)<br />

• Patients with<br />

oropharyngeal<br />

candidiasis<br />

• Non-neutropenic<br />

cancer patients<br />

Dermatophyte toenail<br />

onychomycosis<br />

n=279 Fluconazole vs.<br />

itraconazole<br />

- Terbinafine,<br />

itraconazole,<br />

fluconazole, and<br />

• Of the patients treated with voriconazole, 94.8% (n=108) exhibited<br />

endoscopically proven cure, compared with 90.1% (n=127) in the<br />

fluconazole group.<br />

• Voriconazole (200mg BID) is at least as effective as fluconazole<br />

(200mg QD) in the treatment of microbiologically and histologically<br />

proven esophageal candidiasis in immunocompromised patients,<br />

including those with severe AIDS and a baseline CD4 count of 3 times the upper limit of the normal range in aspartate<br />

transaminase (20% vs. 8%), alanine transaminase (11% vs. 7%) and<br />

alkaline phosphatase (10% vs. 8%) were more frequently observed in<br />

the voriconazole group than in the fluconazole group.<br />

• Seven patients (3.5%) in the voriconazole group and 2 patients (1.1%)<br />

taking fluconazole discontinued because of lab abnormalities.<br />

- In evaluating the efficacy, safety, and tolerance of fluconazole and itraconazole in<br />

patients with oropharyngeal candidiasis:<br />

• The clinical cure rate was 74% for fluconazole and 62% for<br />

itraconazole (p=0.04, 95% Confidence Interval (CI): 0.5-23.3%).<br />

• The mycological cure rate was 80% for fluconazole and 68% for<br />

itraconazole (p=0.03, 95% CI: 1.2-22.6%).<br />

• The safety and tolerance profile of both drugs were comparable.<br />

• Summary: In patients with cancer and oropharyngeal candidiasis,<br />

fluconazole has a significantly better clinical and mycological cure rate<br />

All studies<br />

included in the<br />

meta-analysis<br />

compared with itraconazole.<br />

In reviewing the relevant studies with the oral antifungals for the treatment of<br />

onychomycosis:<br />

• The change in efficacy of mycological cure rates from the first trial to<br />

75


Marr KA, et<br />

al. 56<br />

Shikanai-<br />

Yasuda MA,<br />

et al. 57<br />

randomized<br />

controlled<br />

trials<br />

Randomized<br />

trial<br />

Randomized<br />

trial<br />

Patients receiving<br />

allogeneic stem cell<br />

transplants.<br />

Patients with active<br />

paracoccidioidomycosis<br />

griseofulvin<br />

n=304 Fluconazole<br />

400mg/day<br />

vs.<br />

itraconazole oral<br />

solution 2.5mg/kg<br />

TID vs.<br />

intravenous<br />

itraconazole<br />

200mg QD<br />

n=28 Itraconazole 50-<br />

100mg day,<br />

ketoconazole<br />

200-400 mg day,<br />

or<br />

sulfadiazine 100-<br />

had to include<br />

standard dosing,<br />

duration and<br />

follow-up<br />

180 days post<br />

stem cell<br />

transplant, or<br />

until 4 weeks<br />

after<br />

discontinuation<br />

of graft-versushost<br />

disease<br />

therapy<br />

the overall cumulative meta-average for each drug comparator is as<br />

follows (with 95% confidence interval): terbinafine, 78 +/- 6% (n = 2<br />

studies, 79 patients) to 76 +/- 3% (n = 18 studies, 993 patients) (p =<br />

0.68); itraconazole pulse, 75 +/- 10% (n = 1 study, 20 patients) to 63<br />

+/- 7% (n = 6 studies, 318 patients) (p = 0.25); itraconazole<br />

continuous, 63 +/- 5% (n = 1 study, 84 patients) to 59 +/- 5% (n = 7<br />

studies, 1131 patients) (p = 0.47); fluconazole, 53 +/- 6% (n = 1 study,<br />

72 patients) to 48 +/- 5% (n = 3 studies, 131 patients) (P = 0.50); and<br />

griseofulvin, 55 +/- 8% (n = 2 studies, 109 patients) to 60 +/- 6% (n =<br />

3 studies, 167 patients) (p = 0.41).<br />

• The cumulative meta-analytical average of mycological cure rates<br />

when comparing RCTs vs. open studies was: terbinafine, 76 +/- 3% (n<br />

= 18 studies, 993 patients) vs. 83 +/- 12% (n = 2 studies, 391 patients)<br />

(p = 0.0028); itraconazole pulse, 63 +/- 7% (n = 6 studies, 318<br />

patients) vs. 84 +/- 9% (n = 3 studies, 194 patients) (p = 0.0001); and<br />

fluconazole, 48 +/- 5% (n = 3 studies, 131 patients) vs. 79 +/- 3% (n =<br />

3 studies, 208 patients) (p = 0.0001).<br />

In evaluating fluconazole and itraconazole in the prevention of invasive mold<br />

infections:<br />

• More patients in the itraconazole arm developed hepatotoxicities, and<br />

more patients were discontinued from itraconazole because of<br />

toxicities or gastrointestinal (GI) intolerance (36% versus 16%, p<br />


Bhogal CS,<br />

et al. 58<br />

Shang H, et<br />

al. 59<br />

Bernhardt J,<br />

et al. 60<br />

Randomized<br />

trial<br />

Double blind,<br />

randomized,<br />

multi-center<br />

study<br />

In vitro<br />

efficacy of<br />

voriconazole<br />

and<br />

fluconazole on<br />

multilayered<br />

esophageal<br />

epithelium<br />

• Patients with<br />

moderate to<br />

extensive pityriasis<br />

versicolor<br />

• Confirmation of the<br />

disease by KOH and<br />

Wood’s lamp<br />

examination<br />

Patients with deep and<br />

shallow fungal infections<br />

• Esophageal<br />

epithelium was<br />

infected with<br />

Candida albicans<br />

(SC5314)<br />

• The fluconazole<br />

group was also<br />

exposed to other<br />

clinical strains of<br />

150mg/kg/day up<br />

to 6grams/day<br />

n=180 Ketoconazole<br />

200mg QD<br />

(Category II) x 10<br />

days,<br />

ketoconazole<br />

400mg single<br />

dose (Category I),<br />

fluconazole<br />

400mg single<br />

dose (Category<br />

III), or<br />

fluconazole<br />

150mg per week<br />

for 4 weeks<br />

(Category IV)<br />

Follow-up was<br />

done at 2 and 4<br />

weeks, and then<br />

at 3, 6, and 12<br />

months after<br />

treatment<br />

n=222 Fluconazole 200-400mg QD x 1-8<br />

weeks, fluconazole 150mg single<br />

dose, ketoconazole 400mg QD for 1-<br />

8 weeks for fungal disease and 5<br />

days in patients with fungal vaginitis<br />

- Fluconazole<br />

vs.<br />

voriconazole<br />

Voriconazole<br />

• The test of the hypothesis that the drugs reduced antibody levels up to<br />

ten months of treatment showed a p value of p=0.0001 for itraconazole,<br />

0.017 for ketoconazole, and 0.0012 for sulfadiazine, a reduction that<br />

was similar for each group.<br />

• Summary: This study did not show superiority of any one regimen<br />

over the others in the clinical and serological responses of patients with<br />

moderately severe disease.<br />

In evaluating single vs. multiple doses of ketoconazole and fluconazole for the<br />

treatment of pityriasis versicolor:<br />

• After four weeks of treatment, clinical cure was observed in 66.6%<br />

(Category I), 73.3% (Category II), 80% (Category III) and 59.9%<br />

(Category IV) of patients.<br />

• Mycological cure after four weeks of treatment was observed in 53.3%<br />

(Category I), 73.3% (Category II), 82.2% (Category III) and 64.4%<br />

(Category IV) of patients.<br />

• After twelve months of follow-up, maximum relapses were observed<br />

with Category I. No relapse was seen in Category III patients. The time<br />

period of relapse varied from three to ten months.<br />

• Summary: Single dose 400 mg oral fluconazole provides the best<br />

clinical as well as mycological cure rate with no relapse during twelve<br />

months of follow-up.<br />

In comparing the cure and eradication rates of fluconazole vs. ketoconazole in the<br />

treatment of various fungal infections:<br />

• The cure rate in the fluconazole group was 81.3% (52/64), while the<br />

cure rate in group B was 58% (35/60) (p0.05).<br />

• There were mild and transient vomiting, nausea, and anorexia in the<br />

study. In the ketoconazole group, there was one case of ALT elevation<br />

in each group (p


Lipozencic Double blind,<br />

J, et al. 61 randomized,<br />

parallel-group,<br />

multi-center,<br />

durationfinding<br />

study<br />

Candida. distinct. Fluconazole was more effective against clinical strains of C.<br />

albicans than against the type strain.<br />

Griseofulvin<br />

Pediatric patients with<br />

tinea capitis<br />

n=134 Terbinafine for either 6, 8, 10, or 12<br />

weeks, vs. 12 weeks of high-dose<br />

griseofulvin<br />

In evaluating the efficacy of terbinafine vs. high-dose griseofulvin for<br />

tinea capitis:<br />

• Effective treatment was observed at the end of the study in 62% of<br />

patients treated with terbinafine for six weeks and in 63% treated for 8<br />

weeks. Mycological cure was obtained in 59% and 57%, respectively,<br />

and clinical cure in 76% and 80%.<br />

• In the griseofulvin group, effective treatment was 88%, mycological<br />

cure was 76% and clinical cure 96%. However, these high rates were<br />

believed to be due to the high dosage of this drug and the prolonged<br />

course of treatment.<br />

• Complete cure was observed at the end of study in 62% patients<br />

treated with terbinafine for 6 weeks, in 60% treated for 8 weeks and in<br />

84% patients treated with griseofulvin for 12 weeks.<br />

Wingfield<br />

AB, et al. 62<br />

Randomized<br />

trial<br />

• Age 12-76<br />

• Diagnosis of tinea<br />

imbricata<br />

Miscellaneous<br />

n=59 Griseofulvin 500mg BID for 4<br />

weeks, terbinafine 250mg QD for 4<br />

weeks, itraconazole 200mg BID for 1<br />

week, or fluconazole 200mg once<br />

weekly for 4 weeks<br />

In evaluating the efficacy of griseofulvin, terbinafine, itraconazole, and<br />

fluconazole in the treatment of tinea imbricata:<br />

• Significant remission was achieved in the terbinafine and griseofulvin<br />

groups, lasting up to eight weeks after cessation of therapy.<br />

• The fluconazole group experienced no significant remission, and<br />

remission was of short duration in the itraconazole group.<br />

• No adverse events were reported, and non-compliance with<br />

medications or follow-up was the only reason for removal from the<br />

study.<br />

• Summary: Griseofulvin and terbinafine are effective in the treatment<br />

of TI. The decision of whether to treat at all and which medication to<br />

choose depends greatly on the extent of involvement, the social<br />

situation, and the availability of resources such as laboratory testing<br />

and follow-up.<br />

78


Gupta AK,<br />

et al. 63<br />

Pfaller MA,<br />

et al. 64<br />

Prospective,<br />

comparative,<br />

single-blinded,<br />

randomized,<br />

parallel-group<br />

study<br />

Susceptibility<br />

testing of<br />

isolates of<br />

Candida<br />

glabrata by<br />

geographical<br />

location<br />

Moderate to severe<br />

onychomycosis of the<br />

toenails caused by S.<br />

brevicaulis<br />

601 invasive isolates of<br />

Candida glabrata<br />

n=59 Griseofulvin 600mg BID for 12<br />

months, ketoconazole 200mg QD for<br />

4 months, itraconazole pulse for 3<br />

pulses with each pulse at a dose of<br />

200mg BID for 1 week with 3 weeks<br />

off between pulses, fluconazole<br />

150mg QD for 12 weeks, or<br />

terbinafine 250mg QD for 12 weeks<br />

- Amphotericin B,<br />

flucytosine,<br />

fluconazole,<br />

voriconazole, and<br />

caspofungin<br />

In comparing the efficacy and tolerability of 5 different oral antifungal agents in<br />

the treatment of S. brevicaulis onychomycosis:<br />

• At month 12 after the start of treatment, the response was:<br />

griseofulvin, CC 3/11, MC 0/11, CC + MC 0/11; ketoconazole, CC<br />

10/12, MC 8/12, CC + MC 8/12; itraconazole, CC 12/12, MC 12/12,<br />

CC + MC 12/12; terbinafine, CC 12/12, MC 11/12, CC + MC 11/12,<br />

and fluconazole, CC 8/12, MC 8/12, CC + MC 8/12. CC is clinical<br />

cure and MC is mycological cure.<br />

• Adverse effects consisted of: griseofulvin, gastro-intestinal symptoms,<br />

allergic reaction, photodermatitis, hepatic and renal dysfunction in 11<br />

patients with discontinuation of treatment in three patients;<br />

ketoconazole, hepatic dysfunction but no symptomatic changes in two<br />

patients; itraconazole, nausea and vomiting in two patients;<br />

terbinafine, taste disturbance in two patients, nausea in three patients,<br />

and fluconazole, severe gastro-intestinal events in five patients. None<br />

of the patients receiving ketoconazole, itraconazole, terbinafine or<br />

fluconazole discontinued treatment.<br />

• Summary: Itraconazole and terbinafine demonstrate efficacy against<br />

some cases of S. brevicaulis toe onychomycosis. These agents also<br />

appear to be safe in the course of therapy for toe onychomycosis.<br />

Griseofulvin is ineffective against toe onychomycosis caused by S.<br />

brevicaulis. Ketoconazole is not recommended for toe onychomycosis<br />

given its potential for adverse effects, particularly with the availability<br />

of the newer antifungal agents.<br />

- In evaluating the susceptibilities of 601 invasive isolates of Candida glabrata to 5<br />

antifungal agents:<br />

• Caspofungin (MIC at which 90% of isolates tested are susceptible<br />

[MIC(90)], 0.12 microg/ml; 100% of strains are susceptible [S] at a<br />

MIC of


Silling, et<br />

al. 65<br />

Prospective<br />

study<br />

Patients with neutropenia<br />

secondary to<br />

hematological<br />

malignancies<br />

n=98 Fluconazole<br />

vs.<br />

amphotericin B<br />

plus flucytosine<br />

If no response<br />

to fluconazole<br />

after day 7,<br />

patients were<br />

switched to the<br />

amphotericin<br />

B/flucytosine<br />

group<br />

In comparing the efficacy and tolerability of fluconazole (FCA) to that of<br />

amphotericin B plus flucytosine (ABF) in neutropenic patients:<br />

• Overall in 44/51 (86.2%) of the fluconazole and 37/47 (78.8%) of the<br />

amphotericin B/flucytosine group defervescence was observed.<br />

• 46 patients (21 FCA, 25 ABF) developed radiological signs of<br />

pneumonia. Resolution of infiltrates was achieved in 5/21 FCA and<br />

20/25 ABF patients, and another 10 of 15 initially not responding<br />

patients showed regression when switched to ABF, 5 of these had<br />

highly suspected aspergillosis.<br />

• Adverse events occurred in 19.6% of FCA and 97.9% of ABF patients.<br />

• Summary: Fluconazole and amphotericin B/flucytosine seem to be<br />

equally effective. In view of its lower toxicity fluconazole may be<br />

preferred as first line empiric antifungal agent, but in case of<br />

nonresponse, pneumonia or aspergillosis it may be replaced by<br />

amphotericin B combined with flucytosine.<br />

80


Additional Evidence<br />

Dose Simplification: A majority of the antifungals in this class are dosed once or twice daily.<br />

The exceptions to this include flucytosine (Q6 hours) and nystatin (Q6-Q8 hours). Additionally,<br />

antifungals are indicated primarily for acute use and are routinely not continued as maintenance<br />

therapy. Injectable antifungal therapies are routinely given to hospitalized or skilled nursing<br />

patients with more severe disease, who are under direct supervision.<br />

Although no studies were found in a literature search of Medline or Ovid that specifically looked<br />

at compliance with antifungal therapies, one study reported one of the biggest problems associated<br />

with the treatment of onychomycosis with oral antifungal agents is compliance with long-term<br />

therapy. 66<br />

Stable Therapy: Patients started on one of the antifungals in this class should continue on that<br />

drug until completion of the course of therapy, unless the patient does not respond to the therapy<br />

or is intolerant due to adverse events. Choice of treatment should also coincide with identification<br />

of the fungus and susceptibility testing of the organism. Antifungal resistance should also be a<br />

consideration. Because antifungal therapy is acute and continuation is determined by positive<br />

response, switching between antifungal agents is directly related to treatment failure and drug<br />

intolerance.<br />

To evaluate antifungal drug resistance, 1,143 cases of Candida bloodstream infections were<br />

identified in the Baltimore, Maryland and Connecticut areas. 67 Candida albicans comprised 45%<br />

of the isolates, followed by C. glabrata (24%), C. parapsilosis (13%), and C. tropicalis (12%).<br />

Only 1.2% of C. albicans isolates were resistant to fluconazole (MIC, > or = 64 microg/ml),<br />

compared to 7% of C. glabrata isolates and 6% of C. tropicalis isolates. Only 0.9% of C. albicans<br />

isolates were resistant to itraconazole (MIC, > or = 1 micro g/ml), compared to 19.5% of C.<br />

glabrata isolates and 6% of C. tropicalis isolates. Only 4.3% of C. albicans isolates were resistant<br />

to flucytosine (MIC, > or = 32 microg/ml), compared to < 1% of C. parapsilosis and C. tropicalis<br />

isolates and no C. glabrata isolates. Another study evaluated the prevalence and fluconazole<br />

susceptibility of Candida species in healthcare workers and non-healthcare workers and found that<br />

there was no significant difference in prevalence or species of oropharyngeal Candida and<br />

fluconazole resistance was infrequent. 68<br />

The mechanism of fluconazole resistance to C. glabrata appears to involve cellular resistance and<br />

increased drug efflux as measured by decreased accumulation of fluconazole and increased<br />

abundance of transcripts from two drug transporters. 69<br />

One case report presented data of a successful treatment of fluconazole resistant Candida with the<br />

combination of fluconazole plus flucytosine. 70 The addition of flucytosine to the treatment<br />

regimen in this case showed an additive interaction of fluconazole and flucytosine.<br />

A study evaluated the in vitro activities of multiple antifungal therapies against non-albicans<br />

bloodstream yeast infections. 71 For amphotericin B, the end-point was defined as the minimal<br />

antifungal concentration that exerts 90% inhibition compared with the control well growth. For<br />

the azoles, the end-points were determined at 50% inhibition of growth. Amphotericin B is highly<br />

active with 97% of isolates inhibited by < or =1 microg ml(-1). Decreased susceptibility or<br />

resistance to fluconazole was the rule among C. krusei, which is intrinsically resistant to<br />

fluconazole. For C. glabrata isolates, resistance to fluconazole and itraconazole was measured in<br />

13% and 17% of the isolates respectively. Voriconazole was quite active in vitro against all the<br />

isolates with a MIC90% of < or =1 microg ml(-1).<br />

Multiple clinical studies support use of the lipid amphotericin B formulations after a trial of<br />

conventional amphotericin B, largely due to differences from significantly higher renal toxicity<br />

with conventional amphotericin B. One study suggested amphotericin B is more effective than<br />

fluconazole in candidemias caused by some non-albicans Candida species. 39 Additionally,<br />

caspofungin has been shown to be effective in patients refractory to other antifungal therapy, and<br />

in combination with other antifungals. Voriconazole is an oral option that may also be used in<br />

combination with other antifungal treatments.


Impact on Physician Visits: Multiple pharmacoeconomic studies have looked at the treatments<br />

for onychomycosis. However, data is reported in terms of cost-savings, not in physician visits,<br />

and therefore cannot be included in this review. This section is not relevant to the injectable<br />

medications likely given during hospitalizations. No further data was found in a literature search<br />

of Medline and Ovid pertaining to antifungal use and physician visits.<br />

IX.<br />

Conclusions<br />

1) The lipid amphotericin B formulations are indicated for use for various fungal infections and in<br />

most cases, when renal toxicities/impairment or adverse events preclude the use of conventional<br />

amphotericin B. Conventional amphotericin B is available in a generic formulation, while there is<br />

not a generic lipid amphotericin formulation available. While studies have shown little differences<br />

in efficacy between the lipid-based formulations and conventional amphotericin B, limited clinical<br />

studies have directly compared the lipid amphotericin B agents. Clinical evidence that is available<br />

indicates the lipid amphotericin B agents are comparatively effective for the treatment of<br />

suspected or documented fungal infections. The amphotericin B agents offer benefits to patients<br />

with life-threatening fungal infections. As a result, there is not a role for these agents in general<br />

use. Because of the limited use in outpatient treatment, the amphotericin B agents should be<br />

available for special needs/circumstances that require medical authorization through the prior<br />

authorization process. Therefore, the brand amphotericin B agents within the class reviewed are<br />

comparable to each other and to the generics in the class and offer no significant advantage over<br />

other alternatives in general use.<br />

2) Some clinical comparative data is available for the other antifungals in this class, focusing on<br />

comparative studies for specific indications. Clinical evidence suggests greater efficacy<br />

(mycological cure) of terbinafine compared with itraconazole in the treatment of onychomycosis.<br />

However, treatment of onychomycosis is cosmetic in most cases, and therapy should be reserved<br />

for patients who are predisposed to foot complications (e.g. patients with diabetes). This will<br />

allow patients who medically need these agents, to receive them. As a result, there is not a role for<br />

the antifungal oncyomycosis agents in general use. Additionally, the following antifungal agents<br />

are available in a generic formulation: fluconazole (oral and IV), ketoconazole, nystatin, and<br />

griseofulvin (ultramicrosize). Therefore, all brand products within the class reviewed are<br />

comparable to each other and to the generics in the class and offer no significant clinical<br />

advantage over other alternatives in general use.<br />

3) The remaining agents, caspofungin, flucytosine, IV itraconazole, and voriconazole have<br />

indications for serious, invasive infections, and use of voriconazole and caspofungin is indicated<br />

in those whose disease is refractory to, or who are intolerant to, other therapies. In life-threatening<br />

illness, combinations of these anti-fungals and other antifungals already mentioned may be used.<br />

As caspofungin and voriconazole are newer agents, little direct comparative efficacy data is<br />

available to evaluate their use. These drugs should be made available with medical justification<br />

through the prior authorization process. Therefore, the brands of caspofungin, flucytosine, IV<br />

itraconazole and voriconazole are comparable to each other and to the generics and OTC products<br />

in this class and offer no significant clinical advantage over other alternatives in general use.<br />

X. Recommendations<br />

No brand antifungal is recommended for preferred status.<br />

82


References<br />

1. Merck & Co. Introduction to medical mycology. Whitehouse Station, New Jersey: North Wales<br />

Press, Inc; 2001.<br />

2. Pfizer. Diflucan® (fluconazole) prescribing information. New York (NY): 1998.<br />

3. Janssen Pharmaceutica, Inc. Sporanox® (itraconazole) prescribing information. Titusville (NJ):<br />

2002.<br />

4. Janssen Pharmaceutica, Inc. Nizoral® (ketoconazole) prescribing information. Titusville (NJ):<br />

1998.<br />

5. Novartis. Lamisil® (terbinafine) prescribing information. East Hanover (NJ): 2001.<br />

6. Pfizer. Vfend® (voriconazole) prescribing information. New York (NY): 2002.<br />

7. Clinical Pharmacology 2000. [cited 2002 Oct 9] http://cpip.gsm.com/2002.<br />

8. Lacey CF, Armstrong LL, Goldman MP, Lance LL. Drug information handbook. Hudson, Ohio:<br />

Lexi-Comp, Inc; 2001.<br />

9. Dismukes W. Introduction to antifungal drugs. Clin Infect Dis 2000;3: 653-657.<br />

10. Sobel JD for the mycoses study group. Practice guidelines for the treatment of fungal infections.<br />

Clin Infect Dis 2000; 30:652.<br />

11. Galgiani JN, Ampel NM, Catanzaro A, et al. Practice guidelines for the treatment of<br />

coccidioidomycosis. Clin Infect Dis 2000;30: 658-61.<br />

12. Kauffman CA, Hajjeh R & Chapman W. Practice guidelines for the management of patients with<br />

sporotrichosis. Clin Infect Dis 2000;30 684-87.<br />

13. Wheat J, Sarosi G, McKinsey D et al. Practice guidelines for the management of patients with<br />

histoplasmosis. Clin Infect Dis 2000;30: 688-95.<br />

14. Chapman SW, Bradsher RW, Campbell D et al. Practice guidelines for the management of<br />

patients with blastomycosis. Clin Infect Dis 2000;30: 679-83.<br />

15. Saag MS, Graybill RG, Larsen RA et al. Practice guidelines for the management of cryptococcal<br />

disease. Clin Infect Dis 2000;30: 710-18.<br />

16. Stevens DA, Kan VL, Judson MA et al. Practice guidelines for diseases caused by aspergillus.<br />

Clin Infect Dis 2000;30: 696-709.<br />

17. Rex JH, Walsh TJ, Sobel JD et al. Practice guidelines for the treatment of candidiasis. Clin Infect<br />

Dis 2000;30: 662-78.<br />

18. Pappas PG, Rex JH, Sobel JD, et al. Guidelines for treatment of candidiasis. Clin Infect Dis Jan<br />

15 2004;38(2):161-89.<br />

19. Pappas PG, Rex JH, sobel JD, et al. National Guideline Clearinghouse. Guidelines for the<br />

treatment of candidiasis. Available at: www.guideline.gov. Accessed July 26, 2004.<br />

20. Rodgers P, Bassler M. Treating Onychomycosis. Am Fam Physician Feb 15 2001;63(4):663-672.<br />

21. University of Texas at Austin, School of Nursing, Family Nurse Practitioner Program.<br />

Recommendations for the management of onychomycosis in adults. Austin (TX):University of<br />

Texas at Austin, School of Nursing; May 16 2003: 16.<br />

22. University of Texas at Austin, School of Nursing, Family Nurse Practitioner Program. National<br />

Guideline Clearinghouse. Recommendations for the management of onychomycosis in adults.<br />

Available at: www.guideline.gov. Accessed July 26, 2004.<br />

23. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58,<br />

2004. Thomson PDR. Montvale, NJ. 2004.<br />

24. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

25. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

26. Bellman R, Egger P, Wiedermann CJ. Differences in pharmacokinetics of amphotericin B lipid<br />

formulations despite clinical equivalence. Clin Inf Dis 2003;36:1500-1501.<br />

27. Dupont B. Overview of the lipid formulations of amphotericin B. J Antimicrob Chemother 2002<br />

Feb;49 Suppl 1:31-6.<br />

28. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

29. Rosemurgy AS, Markowsky S, Goode SE, et al. Bioavailability of fluconazole in surgical<br />

intensive care unit patients: a study comparing routes of administration. J Trauma<br />

1995;39(3):445-447.<br />

30. Nicolau DP, Crowe H, Nightingale CH, et al. Bioavailability of fluconazole administered via a<br />

feeding tube in intensive care patients. J Antimicrob Chemother 1995;36(2):395-401.<br />

83


31. Pelz RK, Lipsett PA, Swoboda SM, et al. Enteral fluconazole is well absorbed in critically ill<br />

surgical patients. Surgery 2002;131:534-540.<br />

32. Buijk SLC, Gyssens IC, Mouton JW, et al. Pharmacokinetics of sequential intravenous and<br />

enteral fluconazole in critically ill surgical patients with invasive mycoses and compromised<br />

gastro-intestinal function. Intensive Care Med;27:115-121.<br />

33. Tosti A, Piraccini BM, Iorizzo M, et al. Management of onychomycosis in children. Dermatol<br />

Clin 2003;21(3):507-509.<br />

34. Wong-Beringer A, Jacobs RA, Guglielmo BJ. Lipid formulations of amphotericin B: clinical<br />

efficacy and toxicities. Clin Infect Dis 1998 Sep;27(3):603-18.<br />

35. Miller CB, Waller EK, Klingemann HG, et al. Lipid formulations of amphotericin B preserve and<br />

stabilize renal function in HSCT recipients. Bone Marrow Transplant 2004 Mar;33(5):543-8.<br />

36. Fleming RV, Kantarjian HM, Husni R, et al. Comparison of amphotericin B lipid complex<br />

(ABLC) vs. AmBisome in the treatment of suspected or documented fungal infections in patients<br />

with leukemia. Leuk Lymphoma 2001 Feb;40(5-6):511-20.<br />

37. Cagnoni PJ. Liposomal amphotericin B versus conventional amphotericin B in the empirical<br />

treatment of persistently febrile neutropenic patients. Antimicrob chemother 2002 Feb;49 Suppl<br />

1:81-6.<br />

38. Subira M, Martino R, Gomez L, et al. Low-dose amphotericin B lipid complex vs. conventional<br />

amphotericin B for empirical antifungal therapy of neutropenic fever in patients with hematologic<br />

malignancies—a randomized, controlled trial. Eur J Haematol 2004 May;72(5):342-7.<br />

39. Kontoyiannis DP, Bodey GP, Mantzoros CS. Fluconazole vs. amphotericin B for the management<br />

of candidaemia in adults: a meta-analysis. Mycoses 2001:44(5):125-35.<br />

40. Johansen HK, Gotzsche PC. Amphotericin B versus fluconazole for controlling fungal infections<br />

in neutropenic cancer patients. Cochrane Database Syst Rev. 2002 (2):CD000239.<br />

41. Bowden R, Chandrasekar P, White MH, et al. A double-blind, randomized, controlled trial of<br />

amphotericin B colloidal dispersion versus amphotericin B for treatment of invasive aspergillosis<br />

in immunocompromised patients. Clin Infect Dis 2002 Aug 15;35(4):359-66.<br />

42. Barrett JP, Vardulaki KA, Conlon C, et al. A systematic review of the antifungal effectiveness<br />

and tolerability of amphotericin B formulations. Clin Ther 2003 May;25(50:1295-320.<br />

43. Wingard JR, White MH, Anaissie E, et al. A randomized, double-blind comparative trial<br />

evaluating the safety of liposomal amphotericin B versus amphotericin B lipid complex in the<br />

empirical treatment of febrile neutropenia. L Amph/ABLC Collaborative Study Group. Clin<br />

Infect Dis 2000 Nov;31(5):1155-63.<br />

44. Clark AD, McKendrick S, Tansey PJ, et al. A comparative analysis of lipid-complexed and<br />

liposomal amphotericin B preparations in haematological oncology. Br J Haematol 1998<br />

Oct;103(1):198-204.<br />

45. Linder N, Klinger G, Shalit I, et al. Treatment of candidaemia in premature infants: comparison<br />

of three amphotericin B preparations. J Antimicrob Chemother 2003 Oct;52(4):663-7.<br />

46. Kartsonis NA, Saah A, Lipka CJ, et al. Second-line therapy with caspofungin for mucosal or<br />

invasive candidiasis: results from the caspofungin compassionate-use study. J Antimicrob<br />

Chemother 2004 May;53(5):878-81.<br />

47. Cesaro S, Toffolutti T, Messina C, et al. Safety and efficacy of caspofungin and liposomal<br />

amphotericin B, followed by voriconazole in young patients affected by refractory invasive<br />

mycosis. Eur J Haematol 2004 Jul;73(1):50-5.<br />

48. Cololbo AL, Perfect J, DiNubile M, et al. Global distribution and outcomes for Candida species<br />

causing invasive candidiasis: results from an international randomized double-blind study of<br />

caspofungin versus amphotericin B for the treatment of invasive candidiasis. Eur J Clin Microbiol<br />

Infect Dis 2003 Aug;22(8):470-4.<br />

49. Mora-Duarte J, Betts R, Rotstein C, et al. Comparison of caspofungin and amphotericin B for<br />

invasive candidiasis. N Engl J Med 2002 Dec 19;347(25):2020-9.<br />

50. Galgiani JN, Catanzaro A, Cloud GA, et al. Comparison of oral fluconazole and itraconazole for<br />

progressive, nonmeningeal coccidioidomycosis. A randomized, double blind trial. Mycoses study<br />

group. Ann Intern Med 2000;133(9): 676-86.<br />

51. Evans EGV, Sigurgeirsson B for the LION study group. Double blind, randomized study of<br />

continuous terbinafine compared with intermittent itraconazole in treatment of toenail<br />

onychomycosis. British Medical Journal 1999;318: 1031-1035.<br />

84


52. Brautigam M, Nolting S, Schopf RE, Weidinger G. Randomized double blind comparison of<br />

terbinafine and itraconazole for treatment of toenail tinea infection. British Medical Journal<br />

1995;311: 919-922.<br />

53. Ally R, Schurmann D, Kreisel W, et al. A randomized, double blind, double-dummy, multicenter<br />

trial of voriconazole and fluconazole in the treatment of esophageal candidiasis in<br />

immunocompromised patients. Clin Infect Dis 2001;33(9): 1447-54.<br />

54. Oude Lashof AM, De Bock R, Herbrecht R, et al. An open multicenter comparative study of the<br />

efficacy, safety, and tolerance of fluconazole and itraconazole in the treatment of cancer patients<br />

with oropharyngeal candidiasis. Eur J Cancer 2004 Jun;40(9):1314-9.<br />

55. Gupta AK, Ryder JE, Johnson AM, et al. Cumulative meta-analysis of systemic antifungal agents<br />

for the treatment of onychomycosis. Br J Dermatol 2004 Mar;150(3):537-44.<br />

56. Marr KA, Crippa F, Leisenring W, et al. Itraconazole versus fluconazole for prevention of fungal<br />

infections in patients receiving allogeneic stem cell transplants. Blood 2004 Feb 15;103(4):1527-<br />

33.<br />

57. Shikanai-Yasuda MA, Benard G, Higaki Y, et al. Randomized trial with itraconazole,<br />

ketoconazole and sulfadiazine in paracoccidioidomycosis. Med Mycol 2002 Aug;40(4):411-7.<br />

58. Bhogal CS, Singal A, Baruah MC. Comparative efficacy of ketoconazole and fluconazole in the<br />

treatment of pityriasis versicolor: a one-year follow-up study. J Dermatol 2001 Oct;28(10):535-9.<br />

59. Shang H, Lu Y, Liang D. Fluconazole versus ketoconazole in systemic fungal infection: a<br />

double-blind randomized study. Zhonghua Nei Ke Za Zhi 1997 May;36(5):317-20.<br />

60. Bernhardt J, Bernhardt H, Knoke M, et al. Influence of voriconazole and fluconazole on<br />

reconstituted multilayered esophageal epithelium infected by Candida albicans. Mycoses 2004<br />

Aug;47(7):330-7.<br />

61. Lipozencic J, Skerlev M, Orofino-Costa R, et al. A randomized, double-blind, parallel-group,<br />

duration-finding study of oral terbinafine and open-label, high-dose griseofulvin in children with<br />

tinea capitis due to Microsporum species. Br J Dermatol 2002 May;146(5):816-23.<br />

62. Wingfield AB, Fernandez-Obregon AC, Wignall FS, et al. Treatment of tinea imbricata: a<br />

randomized clinical trial using griseofulvin, terbinafine, itraconazole, and fluconazole. Br J<br />

Dermatol 2004 Jan;150(1):119-26.<br />

63. Gupta AK, Gregurek-Novak T. Efficacy of itraconazole, terbinafine, fluconazole, griseofulvin,<br />

and ketoconazole in the treatment of Scopulariopsis brevicaulis causing onychomycosis of the<br />

toes. Dermatology 2001:202(3):235-8.<br />

64. Pfaller MA, Messer SA, Boyken L, et al. Geographic variation in the susceptibilities of invasive<br />

isolates of Candida glabrata to seven systemically active antifungal agents: a global assessment<br />

from the ARTEMIS Antifungal Surveillance Program conducted in 2001 and 2002. J Clin<br />

Microbiol 2004 Jul;42(7):3142-6.<br />

65. Silling G, Fegeler W, Roos N, et al. Early empiric antifungal therapy of infections in neutropenic<br />

patients comparing fluconazole with amphotericin B/flucytosine. Mycoses 1999;42 Suppl 2:101-<br />

4.<br />

66. Iozumi K, Hattori N, Adachi M, et al. Long-term follow-up study of onychomycosis: cure rate<br />

and dropout rate with oral antifungal treatments. J Dermatol 2001 Mar;28(3):128-36.<br />

67. Hajjeh RA, Sofair AN, Harrison LH, et al. Incidence of bloodstream infections due to Candida<br />

species and in vitro susceptibilities of isolates collected from 1998 to 2000 in a population-based<br />

active surveillance program. J Clin Microbiol 2004 Apr;42(4):1519-27.<br />

68. Klein JD, Levin S. Prevalence of fluconazole susceptibility of Candida species recovered from the<br />

oropharynx of healthcare workers and non healthcare workers. Infect Control Hosp Epidemiol<br />

2004 Apr;25(4):352-4.<br />

69. Bennett JE, Izumikawa K, Marr KA. Mechanism of increased fluconazole resistance in Candida<br />

glabrata during prophylaxis. Antimicrob Agents Chemother 2004 May;48(5):1773-7.<br />

70. Girmenia C, Venditti M, Martino P, et al. Fluconazole in combination with flucytosine in the<br />

treatment of fluconazole-resistant Candida infections. Diagn Microbiol Infect Dis 2003<br />

Jul;46(3):227-31.<br />

71. Swinne D, Watelle M, Van der Flaes M, et al. In vitro activities of voriconazole (UK-109, 496),<br />

fluconazole, itraconazole, and amphotericin B against 132 non-albicans bloodstream yeast isolates<br />

(CANARI study). Mycoses 2004 Jun;47(5-6):177-83.<br />

85


I. Overview<br />

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Cephalosporins<br />

AHFS 081206<br />

October 27, 2004<br />

Cephalosporins are commonly used antibiotics in the ambulatory and hospital setting for both<br />

adults and children due to low toxicity and activity against various organisms. Twenty-five<br />

cephalosporins are currently available in the United States. Cephalosporins are grouped into<br />

"generations" according to spectrum of activity. First-generation cephalosporins are most active<br />

against gram-positive aerobes, while third-generation drugs are most active against gram-negative<br />

aerobes.<br />

All first generation oral cephalosporins are available as generic products. Only two of the oral<br />

second generation products are availably generically, cefaclor and cefuroxime. For the third<br />

generation oral agents, only the generic version of Vantin tablets are available. Another third<br />

generation cephalosporin, Suprax, was discontinued by the manufacturer. The future availability<br />

of Suprax is unclear.<br />

Based on microbiological spectrum and clinical indications, the brand name products do not offer<br />

additional benefit over the existing generics. Oral third generation agents are inactive against<br />

Enterobacter, Pseudomonas and most of the anaerobic organisms. Among the third generation<br />

oral agents, cefixime and ceftibuten are inactive against Staphylococci. Cefditoren is the newest<br />

addition to this class and has increased activity against gram-positive organisms. All of the oral<br />

third generation agents, except cefditoren, are available as suspensions. In clinical trials, third<br />

generation oral agents demonstrated marginal benefit in clinical outcomes when compared to oral<br />

first or second generation agents in the treatment of mild to moderate respiratory, urinary tract,<br />

skin and structure infections. However, third generation oral agents have improved<br />

microbiological eradication rates and organisms develop resistance at slower rates. This review<br />

encompasses all dosage forms and strengths. Table 1 lists the drugs included in this review.<br />

Table 1. Cephalosporins in this <strong>Review</strong><br />

Generic Name Generation Formulation Example Brand Name (s)<br />

Cefadroxil First Tablet, Capsule, Oral Suspension Duricef*<br />

Cefazolin Injection Ancef*, Kefzol<br />

Cephalexin Tablet, Capsule, Oral Suspension Keflex*, Panixine<br />

Cephapirin Injection Cefadyl<br />

Cephradine<br />

Capsule, Oral Suspension, Injection<br />

Anspor*, Velosef*<br />

Cefaclor Second Tablet, Capsule, Oral Suspension Ceclor*, Ceclor CD, Raniclor<br />

Cefprozil Tablet, Oral Suspension Cefzil<br />

Cefamandole -- Mandol†<br />

Cefuroxime<br />

Tablet, Oral Suspension, Injection Ceftin*, Kefurox, Alti, Zinacef<br />

Cefdinir Third<br />

Capsule, Oral Suspension Omnicef<br />

Cefditoren Tablet Spectracef<br />

Cefepime Injection Maxipime<br />

Cefixime Tablet, Oral Suspension Suprax†<br />

Cefotaxime Injection Claforan*, Claforan Galaxy<br />

Cefpodoxime Tablet, Oral suspension Vantin* (tablets only generic)<br />

Ceftazidime Injection Ceptaz, Fortaz, Tazicef<br />

Ceftibuten Capsule, Oral Suspension Cedax<br />

Ceftizoxime Injection Cefizox<br />

Ceftriaxone<br />

Injection<br />

Rocephin<br />

*Generic Available.<br />

†This product has been discontinued and is no longer marketed in the United States.<br />

86


II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

The cephalosporins are indicated for a wide range of indications, as detailed in the following<br />

section. These drugs play an important role in the treatment guidelines for many infections<br />

diseases. Of importance are the updated (March 2004) treatment guidelines for the diagnosis and<br />

management of acute otitis media. The guidelines are supported by the American Academy of<br />

Pediatrics and American Academy of Family Physicians. 1<br />

The new guidelines for the treatment of otitis media, middle ear infections, and pain management<br />

associated with both, are based on the age of the child and other factors. Acute otitis media<br />

(AOM) is the most common bacterial illness in children and the one most commonly treated with<br />

antibiotics. 1 There has been significant increase in, and concern about antibacterial resistance of<br />

the organisms that cause AOM. Additionally, the number of office visits for otitis media and<br />

effusion (middle ear fluid, OME) have decreased over the past ten years from 25 million in 1990<br />

to just 16 million in 2000. However, the number of antibiotic prescriptions to treat acute otitis<br />

media has remained constant.<br />

Highlights from the updated guideline include the following recommendations:<br />

1) Accurately diagnose AOM and differentiate it from OME, which requires different<br />

management.<br />

2) Relieve pain, especially in the first 24 hours, with ibuprofen or acetaminophen.<br />

3) Minimize antibiotic side effects by giving parents of select children the option of fighting the<br />

infection on their own for 48-72 hours, then starting antibiotics if they do not improve.<br />

4) Prescribe initial antibiotics for children who are likely to benefit the most from treatment.<br />

5) Encourage families to prevent AOM by reducing risk factors. For babies and infants, these<br />

include breastfeeding for at least six months, avoiding “bottle propping,” and eliminating<br />

exposure to passive tobacco smoke.<br />

6) If antibiotic treatment is agreed upon, the clinician should prescribe amoxicillin for most<br />

children.<br />

The guidelines apply to an otherwise health child with no underlying conditions that would alter<br />

the natural course of AOM. These conditions include, but are not limited to, anatomic<br />

abnormalities such as cleft palate, genetic conditions such as Down syndrome, immune system<br />

disorders, and cochlear implants. Also excluded are children with a clinical recurrence of AOM<br />

within 30 days of AOM with underlying chronic OME.<br />

The following offer other considerations with the use of cephalosporins, with respect to<br />

indications and spectrum of activity:<br />

1. Only one indication, acute otitis media with effusion, has limited evidence for superior<br />

efficacy of one cephalosporin (cefuroxime) over another (cefaclor).<br />

2. Cephalexin is currently the only oral cephalosporin with FDA approval for bone infections<br />

caused by staphylococci or Proteus mirabilis.<br />

3. Cephalexin is currently the only cephalosporin with FDA approval for genitourinary tract<br />

infections including acute prostatitis caused by Escherichia coli, P. mirabilis and Klebsiella<br />

sp.<br />

4. Cefpodoxime is currently the only cephalosporin with FDA approval for ano-rectal infections.<br />

5. Cefuroxime is currently the only cephalosporin with FDA approval for Lyme disease.<br />

87


III.<br />

Comparative Indications of the Cephalosporins<br />

Table 2 lists the indications and organisms covered by the cephalosporin drugs.<br />

Table 2. Indications and Spectrum of the Cephalosporins 2-5<br />

Drug<br />

Indications<br />

Cefadroxil • Escherichia coli<br />

• impetigo<br />

• Klebsiella sp.<br />

• Moraxella catarrhalis<br />

• pharyngitis<br />

• Proteus mirabilis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• urinary tract infection (UTI)<br />

Cefazolin<br />

Cephalexin<br />

• biliary tract infections<br />

• bone and joint infections<br />

• burn wound infection<br />

• endocarditis<br />

• Enterobacter sp.<br />

• epididymitis<br />

• Escherichia coli<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• pneumonia<br />

• prostatitis<br />

• Proteus mirabilis<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• surgical infection prophylaxis<br />

• urinary tract infection (UTI)<br />

• prostatitis<br />

• Proteus mirabilis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• urinary tract infection (UTI)<br />

• pneumonia<br />

• bone and joint infections<br />

• cystitis<br />

• Escherichia coli<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

• otitis media<br />

•pharyngitis


Cephapirin • Respiratory tract infections (S. pneumoniae, Klebsiella species, H.<br />

influenzae, S. aureus, group A beta-hemolytic streptococci)<br />

• Skin infections (S. aureus, S. epidermidis, E. coli, P. mirabilis,<br />

Klebsiella species, group A beta-hemolytic streptococci)<br />

• Urinary tract infections (E. coli, Klebsiella species, P. mirabilis, S.<br />

aureus)<br />

• Septicemia (S. viridans, S. aureus, Klebsiella species, E. coli, group<br />

A beta-hemolytic streptococci)<br />

• Endocarditis (S. aureus, S. viridans)<br />

• Osteomyelitis (S. aureus, P. mirabilis, Klebsiella species, group A<br />

beta-hemolytic streptococci)<br />

Cephradine • Escherichia coli<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• otitis media<br />

• pharyngitis<br />

• pneumonia<br />

• prostatitis<br />

• Proteus mirabilis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• urinary tract infection (UTI)<br />

Cefaclor • Propionibacterium acnes<br />

• Proteus mirabilis<br />

• Providencia sp.<br />

• pyelonephritis<br />

• Shigella sp.<br />

• sinusitis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• upper respiratory tract infections<br />

• urinary tract infection (UTI)<br />

• Bacteroides sp.<br />

• Citrobacter diversus<br />

• cystitis<br />

• Escherichia coli<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

• Neisseria gonorrhoeae<br />

• otitis media<br />

• Peptococcus sp.<br />

• Peptostreptococcus sp.<br />

• pharyngitis<br />

• pharyngitis<br />

• pneumonia<br />

89


Cefprozil<br />

Cefuroxime<br />

• Proteus mirabilis<br />

• Salmonella sp.<br />

• Shigella sp.<br />

• sinusitis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Staphylococcus saprophyticus<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus dysgalactiae<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• Vibrio sp.<br />

• Viridans streptococci<br />

• bronchitis<br />

• Citrobacter diversus<br />

• Clostridium perfringens<br />

• Escherichia coli<br />

• Fusobacterium sp.<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

• Neisseria gonorrhoeae<br />

• otitis media<br />

• Peptostreptococcus sp.<br />

• pharyngitis<br />

• pneumonia<br />

• Prevotella melaninogenica<br />

• Propionibacterium acnes<br />

• Actinomyces sp.<br />

• bacteremia<br />

• Bacteroides sp.<br />

• bone and joint infections<br />

• bronchitis<br />

• cellulitis<br />

• cervicitis<br />

• Citrobacter diversus<br />

• Citrobacter freundii<br />

• Clostridium sp.<br />

• Enterobacter aerogenes<br />

• Escherichia coli<br />

• Eubacterium sp.<br />

• Fusobacterium sp.<br />

• gonorrhea<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• impetigo<br />

• Klebsiella pneumoniae<br />

• Klebsiella sp.<br />

• Lactobacillus sp.<br />

• lower respiratory tract infections<br />

• Lyme disease<br />

• meningitis<br />

• Moraxella catarrhalis<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• osteomyelitis<br />

• otitis media<br />

• Peptococcus sp.<br />

90


• Peptostreptococcus sp.<br />

• pharyngitis<br />

• pneumonia<br />

• proctitis<br />

• Propionibacterium sp.<br />

• Proteus mirabilis<br />

• Providencia stuartii<br />

• Salmonella sp.<br />

• septicemia<br />

• Shigella sp.<br />

• sinusitis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• surgical infection prophylaxis<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• urethritis<br />

• urinary tract infection (UTI)<br />

• Veillonella sp.<br />

Cefdinir<br />

Cefditoren<br />

Cefepime<br />

• pneumonia<br />

• Proteus mirabilis<br />

• sinusitis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• Viridans streptococci<br />

• bronchitis<br />

• Citrobacter diversus<br />

• Escherichia coli<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• Klebsiella pneumoniae<br />

• Moraxella catarrhalis<br />

• otitis media<br />

• pharyngitis<br />

• bronchitis<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• Moraxella catarrhalis<br />

• pharyngitis<br />

• pneumonia<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• Acinetobacter sp.<br />

• Aeromonas hydrophila<br />

• bacteremia<br />

• Citrobacter freundii<br />

• Citrobacter koseri<br />

• Enterobacter aerogenes<br />

91


• Enterobacter agglomerans<br />

• Enterobacter cloacae<br />

• Enterobacter sp.<br />

• Escherichia coli<br />

• febrile neutropenia<br />

• Gardnerella vaginalis<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Hafnia alvei<br />

• intraabdominal infections<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

• Morganella morganii<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• pneumonia<br />

• Proteus mirabilis<br />

• Proteus vulgaris<br />

• Providencia rettgeri<br />

• Providencia stuartii<br />

• Pseudomonas aeruginosa<br />

• Salmonella sp.<br />

• Serratia liquefaciens<br />

• Serratia marcescens<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus bovis<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• urinary tract infection (UTI)<br />

• Viridans streptococci<br />

• Yersinia enterocolitica<br />

Cefixime<br />

• Pasteurella multocida<br />

• pharyngitis<br />

• pneumonia<br />

• proctitis<br />

• Proteus mirabilis<br />

• Proteus vulgaris<br />

• Salmonella sp.<br />

• Shigella sp.<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• urethritis<br />

• urinary tract infection (UTI)<br />

• bronchitis<br />

• cervicitis<br />

• Citrobacter amalonaticus<br />

• Citrobacter diversus<br />

• Escherichia coli<br />

• gonorrhea<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• Klebsiella oxytoca<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

92


• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• otitis media<br />

Cefotaxime<br />

Cefpodoxime<br />

• Acinetobacter sp.<br />

• bacteremia<br />

• Bacteroides sp.<br />

• bone and joint infections<br />

• Borrelia burgdorferi<br />

• cervicitis<br />

• Citrobacter sp.<br />

• Clostridium sp.<br />

• Eikenella corrodens<br />

• endometritis<br />

• Enterobacter sp.<br />

• Escherichia coli<br />

• Eubacterium sp.<br />

• gonorrhea<br />

• gynecologic infections<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• intraabdominal infections<br />

• Klebsiella pneumoniae<br />

• Klebsiella sp.<br />

• lower respiratory tract infections<br />

• Lyme disease<br />

• meningitis<br />

• Moraxella catarrhalis<br />

• Morganella morganii<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitides<br />

• pelvic cellulitis<br />

• pelvic inflammatory disease (PID)<br />

• Peptococcus sp.<br />

• Peptostreptococcus sp.<br />

• peritonitis<br />

• pneumonia<br />

• proctitis<br />

• Propionibacterium sp.<br />

• Proteus inconstans<br />

• Proteus mirabilis<br />

• Proteus vulgaris<br />

• Providencia rettgeri<br />

• Providencia stuartii<br />

• Salmonella sp.<br />

• septicemia<br />

• Serratia sp.<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• typhoid fever<br />

• urethritis<br />

• urinary tract infection (UTI)<br />

• ventriculitis<br />

• Yersinia enterocolitica<br />

• pneumonia<br />

• Proteus mirabilis<br />

93


• Proteus vulgaris<br />

• Providencia rettgeri<br />

• sinusitis<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus saprophyticus<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• Streptococcus sp.<br />

• tonsillitis<br />

• upper respiratory tract infections<br />

• urinary tract infection (UTI)<br />

• bronchitis<br />

• Citrobacter diversus<br />

• cystitis<br />

• Escherichia coli<br />

• gonorrhea<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• Klebsiella oxytoca<br />

• Klebsiella pneumoniae<br />

• lower respiratory tract infections<br />

• Moraxella catarrhalis<br />

• Neisseria gonorrhoeae<br />

• otitis media<br />

• Peptostreptococcus magnus<br />

• pharyngitis<br />

Ceftazidime<br />

• Acinetobacter sp.<br />

• bacteremia<br />

• bone and joint infections<br />

• bronchiectasis<br />

• Citrobacter diversus<br />

• Citrobacter freundii<br />

• Clostridium sp.<br />

• Eikenella corrodens<br />

• endometritis<br />

• Enterobacter aerogenes<br />

• Enterobacter agglomerans<br />

• Enterobacter cloacae<br />

• Escherichia coli<br />

• Eubacterium sp.<br />

• febrile neutropenia<br />

• gynecologic infections<br />

• Haemophilus ducreyi<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• intraabdominal infections<br />

• Klebsiella oxytoca<br />

• Klebsiella pneumoniae<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• osteomyelitis<br />

• Pasteurella multocida<br />

• pelvic cellulitis<br />

• Peptococcus sp.<br />

• Peptostreptococcus sp.<br />

• peritonitis<br />

• pneumonia<br />

• Propionibacterium sp.<br />

• Proteus mirabilis<br />

94


Ceftibuten<br />

Ceftizoxime<br />

• Proteus vulgaris<br />

• Providencia rettgeri<br />

• Providencia stuartii<br />

• Pseudomonas aeruginosa<br />

• Salmonella sp.<br />

• septicemia<br />

• Serratia marcescens<br />

• Serratia sp.<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• tonsillitis<br />

• bronchitis<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Moraxella catarrhalis<br />

• otitis media<br />

• pharyngitis<br />

• Acinetobacter sp.<br />

• Actinomyces sp.<br />

• Aeromonas hydrophila<br />

• Bacteroides fragilis<br />

• Bacteroides sp.<br />

• Bifidobacterium sp.<br />

• bone and joint infections<br />

• cervicitis<br />

• Citrobacter freundii<br />

• Citrobacter sp.<br />

• Clostridium perfringens<br />

• Clostridium sp.<br />

• Corynebacterium diphtheriae<br />

• Enterobacter aerogenes<br />

• Enterobacter cloacae<br />

• Enterobacter sp.<br />

• Escherichia coli<br />

• Eubacterium sp.<br />

• Fusobacterium sp.<br />

• gonorrhea<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• intraabdominal abscess<br />

• intraabdominal infections<br />

• Klebsiella pneumoniae<br />

• Klebsiella sp.<br />

• lower respiratory tract infections<br />

• meningitis<br />

• Moraxella catarrhalis<br />

• Moraxella sp.<br />

• Morganella morganii<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• osteomyelitis<br />

• Pasteurella multocida<br />

• pelvic inflammatory disease (PID)<br />

• Peptococcus sp.<br />

• Peptostreptococcus sp.<br />

• peritonitis<br />

• pneumonia<br />

• Propionibacterium sp.<br />

• Proteus mirabilis<br />

• Proteus vulgaris<br />

95


Ceftriaxone<br />

• Providencia sp.<br />

• Providencia stuartii<br />

• Pseudomonas aeruginosa<br />

• Pseudomonas sp.<br />

• Salmonella sp.<br />

• septicemia<br />

• Serratia marcescens<br />

• Serratia sp.<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• Streptococcus sp.<br />

• urethritis<br />

• urinary tract infection (UTI)<br />

• Veillonella sp.<br />

• Yersinia enterocolitica<br />

• Acinetobacter calcoaceticus<br />

• Actinomyces sp.<br />

• bone and joint infections<br />

• Borrelia burgdorferi<br />

• cervicitis<br />

• chancroid<br />

• Citrobacter diversus<br />

• Citrobacter freundii<br />

• Clostridium sp.<br />

• Eikenella corrodens<br />

• endocarditis<br />

• Enterobacter aerogenes<br />

• Enterobacter cloacae<br />

• Escherichia coli<br />

• Fusobacterium sp.<br />

• gonorrhea<br />

• gonorrhea prophylaxis<br />

• gynecologic infections<br />

• Haemophilus ducreyi<br />

• Haemophilus influenzae (beta-lactamase negative)<br />

• Haemophilus influenzae (beta-lactamase positive)<br />

• Haemophilus parainfluenzae<br />

• intraabdominal infections<br />

• Klebsiella oxytoca<br />

• Klebsiella pneumoniae<br />

• Lactobacillus sp.<br />

• lower respiratory tract infections<br />

• meningitis<br />

• Moraxella catarrhalis<br />

• Morganella morganii<br />

• Neisseria gonorrhoeae<br />

• Neisseria meningitidis<br />

• otitis media<br />

• pelvic inflammatory disease (PID)<br />

• Peptococcus sp.<br />

• Peptostreptococcus sp.<br />

• pharyngitis<br />

• pneumonia<br />

• Prevotella melaninogenica<br />

• proctitis<br />

• Propionibacterium sp.<br />

• Proteus mirabilis<br />

• Proteus vulgaris<br />

• Providencia rettgeri<br />

96


• Providencia stuartii<br />

• Salmonella sp.<br />

• septicemia<br />

• Serratia marcescens<br />

• Shigella sp.<br />

• skin and skin structure infections<br />

• Staphylococcus aureus (MSSA)<br />

• Staphylococcus epidermidis<br />

• Streptococcus agalactiae (group B streptococci)<br />

• Streptococcus pneumoniae<br />

• Streptococcus pyogenes (group A beta-hemolytic streptococci)<br />

• surgical infection prophylaxis<br />

• urethritis<br />

• urinary tract infection (UTI)<br />

• Veillonella sp.<br />

• Viridans streptococci<br />

• Yersinia enterocolitica<br />

97


IV.<br />

Pharmacokinetic Parameters of the Cephalosporins<br />

The cephalosporins are usually bactericidal in action. Beta-lactam antibiotics, such as the<br />

penicillins and cephalosporins, bind to bacterial enzymes and inhibit mucopeptide synthesis in the<br />

bacterial cell wall. Table 3 lists the cephalosporin antibiotics and pharmacokinetic parameters for<br />

each.<br />

Table 3. Pharmacokinetic Parameters of the Cephalosporins 2-5<br />

Drug Bioavailability Protein<br />

Binding<br />

Metabolism<br />

Cefadroxil Completely and rapidly 20 No appreciable<br />

absorbed<br />

metabolism.<br />

Cefazolin Administered IV, not well 80-85% Not hepatically<br />

absorbed from the GI tract<br />

metabolized.<br />

Cephalexin Following a 250 or 500mg 10% Largely excreted<br />

oral dose of cephalexin,<br />

unchanged in the<br />

average peak serum<br />

urine.<br />

concentrations of 9 or<br />

15—18mcg/ml,<br />

respectively, are achieved<br />

within 1 hour and mean<br />

serum concentrations<br />

decline to 1.6 or<br />

3.4mcg/ml, respectively,<br />

at 3 hours post-dose.<br />

Active<br />

Metabolites<br />

Elimination Half-Life<br />

- 90% excreted 1-2 hours<br />

unchanged in urine.<br />

- Largely excreted 1-2 hours<br />

unchanged in urine.<br />

- Renal 1 hour<br />

Cephapirin - 44-50 - - 70% recovered<br />

unchanged in urine.<br />

Cephradine Rapidly and almost 8-17% >90% of absorbed - >90 of drug excreted<br />

completely absorbed from<br />

drug is excreted<br />

unchanged in urine.<br />

the GI tract.<br />

unchanged in the<br />

Cefaclor<br />

Readily absorbed from the<br />

GI tract.<br />

urine.<br />

25% Largely excreted<br />

unchanged in the<br />

urine.<br />

Cefprozil Readily absorbed from the<br />

GI tract.<br />

Cefuroxime Peak serum levels of<br />

cefuroxime sodium<br />

(parenteral) occur within<br />

15—60 minutes following<br />

an IM dose. Cefuroxime<br />

axetil is rapidly<br />

hydrolyzed in the<br />

intestinal mucosa, with<br />

37—52% of an oral dose<br />

reaching the systemic<br />

circulation as cefuroxime.<br />

Peak serum levels of<br />

cefuroxime after oral<br />

administration occur<br />

within 2 hours following<br />

an oral dose.<br />

Cefdinir 21% (300mg capsule) 60-70% Not appreciably<br />

16% (600mg capsule)<br />

metabolized.<br />

25% (susp.)<br />

Cefditoren 14% (fasting) 16% (lowfat<br />

88% Minimal<br />

meal)<br />

metabolism.<br />

Cefepime 100% of IM dose 20% 80% recovery of<br />

unchanged drug 8<br />

hours after a dose.<br />

- Largely excreted<br />

unchanged in urine.<br />

36% - - 60% excreted<br />

unchanged in urine.<br />

50% Largely excreted - Largely excreted<br />

unchanged in the<br />

unchanged in urine.<br />

urine.<br />

No<br />

Excretion is primarily<br />

renal.<br />

- Mainly excreted<br />

renally.<br />

- 80% excreted<br />

unchanged in urine.<br />

36 hours<br />

1.3 hours<br />

½-1 hour<br />

1.3 hours<br />

1-2 hours<br />

1.7 hours<br />

1.2 hours<br />

Cefixime 40-50% of oral dose 65% Over 10% of a - 10% excreted in bile. 3-4 hours<br />

2-2.3<br />

hours


Cefotaxime<br />

Cefpodoxime<br />

Ceftazidime<br />

Peak levels following<br />

parenteral administration<br />

occur within 30 minutes<br />

of an IM dose.<br />

Approximately 50% of the<br />

administered dose of<br />

cefpodoxime proxetil is<br />

absorbed systemically.<br />

Administered parenterally;<br />

not absorbed from the GI<br />

dose is excreted in<br />

bile.<br />

30-40% Metabolized<br />

hepatically.<br />

22-33% Minimal<br />

metabolism.<br />

tract.<br />

Ceftibuten 80% relative 65% Minimal<br />

metabolism.<br />

Ceftizoxime<br />

Ceftriaxone<br />

Following IM<br />

administration of 500mg<br />

or 1g of ceftizoxime, peak<br />

ceftizoxime serum<br />

concentrations of 13.7<br />

mcg/ml or 39 mcg/ml,<br />

respectively, occur within<br />

30—60 minutes.<br />

Administered parenterally;<br />

not absorbed from the GI<br />

tract. Peak serum<br />

concentrations of<br />

ceftriaxone occur within<br />

1.5—4 hours following an<br />

IM dose.<br />

Desacetylcefo<br />

taxime<br />

Largely excreted in<br />

urine.<br />

Drug and metabolite<br />

excreted in urine.<br />

- 29-33% excreted<br />

unchanged in urine.<br />

5-24% - - Largely excreted in the<br />

urine.<br />

Cisceftibuten,<br />

transceftibuten<br />

56% excreted in urine<br />

and 39% in feces.<br />

30% Not metabolized. - 80% excreted<br />

85-96% Small amount is<br />

metabolized to<br />

inactive<br />

metabolite.<br />

unchanged in urine.<br />

- 33-67% excreted in<br />

urine and feces.<br />

Drug: 1-<br />

1.5 hours.<br />

Metabolit<br />

e: 1.5-2<br />

hours<br />

3 hours<br />

1.5-2<br />

hours<br />

2-2.5<br />

hours<br />

1.7 hours<br />

5.5-11<br />

hours<br />

99


V. Drug Interactions of the Cephalosporins<br />

Table 4 lists drug interactions of the cephalosporin antibiotics.<br />

Table 4. Drug Interactions of the Cephalosporins 6<br />

Interacting Drug Significance Interaction Mechanism<br />

Amikacin Sulfate,<br />

Gentamicin,<br />

Kanamycin Sulfate,<br />

Neomycin<br />

Netilmicin Sulfate,<br />

Tobramycin<br />

2-Interaction may cause<br />

deterioration in a<br />

patient's clinical status;<br />

occurrence suspected,<br />

established or probable<br />

in well controlled<br />

Nephrotoxicity may be increased.<br />

Bactericidal activity against certain<br />

pathogens may be enhanced.<br />

Interaction appears more prominent<br />

with cephalothin and gentamicin than<br />

with other combinations.<br />

Unknown<br />

Anisindione,<br />

Dicumarol, Warfarin<br />

Sodium<br />

Ethanol<br />

Heparin<br />

Cimetidine,<br />

Famotidine,<br />

Nizatidine, Ranitidine<br />

Colistimethate<br />

studies.<br />

2-Interaction may cause<br />

deterioration in a<br />

patient's clinical status;<br />

occurrence suspected,<br />

established or probable<br />

in well controlled<br />

studies.<br />

2-Interaction may cause<br />

deterioration in a<br />

patient's clinical status;<br />

occurrence suspected,<br />

established or probable<br />

in well controlled<br />

studies.<br />

4-Interaction may cause<br />

moderate-to-major<br />

effects; data are very<br />

limited.<br />

4-Interaction may cause<br />

moderate-to-major<br />

effects; data are very<br />

limited.<br />

4-Interaction may cause<br />

moderate-to-major<br />

effects; data are very<br />

limited.<br />

The anticoagulant effect of warfarin<br />

is increased.<br />

A disulfiram-like reaction manifested<br />

by flushing, tachycardia,<br />

bronchospasm, sweating, nausea and<br />

vomiting may occur when ethanol is<br />

ingested after a patient has taken a<br />

cephalosporin with the<br />

methyltetrazolethiol moiety.<br />

Increased risk of bleeding.<br />

Ranitidine decreases the<br />

bioavailability of certain<br />

cephalosporins.<br />

Coadministration of colistimethate<br />

and cephalothin sodium may increase<br />

the risk of renal dysfunction.<br />

Unknown<br />

Aldehyde dehydrogenase inhibition by<br />

methyltetrazolethiol moiety results in<br />

acetaldehyde accumulation. Occurrence<br />

and severity are unpredictable. Doseresponse<br />

relationships and length of<br />

enzyme inhibition are unknown.<br />

Several cephalosporins have caused<br />

coagulopathies. This might be additive<br />

with heparin.<br />

Unknown. Changes in gastric pH may<br />

affect drug absorption.<br />

Unknown.<br />

100


VI.<br />

Adverse Drug Events of the Cephalosporins<br />

Tables 5 a, b, and c compare adverse events by generation for the drugs in this class.<br />

Table 5a. Common Adverse Events (%) Reported for the Cephalosporins (First Generation)<br />

Adverse Event Cefadroxil Cefazolin Cephalexin Cephapirin Cephradine<br />

Body as a Whole<br />

Information not<br />

Malaise<br />

available<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Epigastric distress<br />

Appetite decrease<br />

Pseudomonas Colitis<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Seizures<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial<br />

Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

a a<br />

a<br />

Hematologic<br />

Neutropenia<br />

Agranulocytosis<br />

a a a a<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

Other<br />

Angioedema<br />

Nephritis<br />

Convulsions<br />

Stevens-Johnson Syndrome<br />

a<br />

a<br />

a- Reported, but percentages not available.<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

101


Table 5b. Common Adverse Events (%) Reported for the Cephalosporins (Second Generation)<br />

Adverse Event Cefaclor Cefprozil Cefuroxime<br />

Body as a Whole<br />

Malaise<br />

a<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Epigastric distress<br />

Appetite decrease<br />

Pseudomonas Colitis<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial<br />

Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

1<br />

3.5/1<br />

2.5<br />

1 in 440<br />

1 in 220<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

a a<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Hematologic<br />

Neutropenia<br />

Leukopenia<br />

Agranulocytosis<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

Other<br />

Angioedema<br />

Convulsions/Seizures<br />

Stevens Johnson Syndrome<br />

a- Reported, but percentages not available.<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

1<br />

a<br />

a<br />

a<br />

0.9<br />

a<br />

a<br />

a<br />

1 in 100<br />

1 in 750<br />

a<br />

a<br />

a<br />

102


Table 5c. Common Adverse Events (%) Reported for the Cephalosporins (Third Generation)<br />

Adverse Event Cefdinir Cefditoren Cefepime Cefixime Cefotaxime<br />

Body as a Whole<br />

Malaise<br />

a<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Epigastric distress<br />

Appetite decrease<br />

Pseudomonas colitis<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial<br />

Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

a<br />

a<br />

a<br />

0.3<br />

a<br />

a<br />

2<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

0.9<br />

a<br />

0.2<br />

a<br />

Hematologic<br />

Neutropenia<br />

Leukopenia<br />

Agranulocytosis<br />

a a<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

Other<br />

Angioedema<br />

Convulsions/Seizures<br />

Stevens-Johnson Syndrome<br />

a<br />

a<br />

a- Reported, but percentages not available.<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

Xerostomia<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

a<br />

Uticaria<br />

a<br />

a<br />

103


Table 5c. (continued) Common Adverse Events (%) Reported for the Cephalosporins (Third Generation)<br />

Adverse Event Cefpodoxime Ceftazidime Ceftibuten Ceftizoxime Ceftriaxone<br />

Body as a Whole<br />

Malaise


VII.<br />

Dosing and Administration for the Cephalosporins<br />

Table 6 details dosing for the cephalosporin antibiotics.<br />

Table 6. Dosing for the Cephalosporins 4<br />

Drug Availability Dose /Frequency/Duration<br />

Cefadroxil Capsules: 500mg<br />

Tablets: 1gram<br />

Powder for oral<br />

suspension:<br />

125mg/5ml<br />

250mg/5ml<br />

500mg/5ml<br />

Cefazolin<br />

Cephalexin<br />

Powder for<br />

Injection:<br />

500mg, 1gm,<br />

10gm, 20gm<br />

Injection:<br />

500mg, 1gm<br />

Tablets/Capsules:<br />

250mg and<br />

500mg<br />

Oral suspension:<br />

125mg/5ml<br />

250mg/5ml<br />

Urinary tract infections: For uncomplicated lower urinary tract infection (e.g., cystitis), the usual<br />

dosage is 1 or 2 g/day in single or 2 divided doses. For all other urinary tract infections, the usual<br />

dosage is 2 g/day in 2 divided doses. Skin and skin structure infections: 1g/day in single or 2<br />

divided doses. Pharyngitis and tonsillitis: Group A B-hemolytic streptococci:<br />

1 g/day in single or 2 divided doses for 10 days. Urinary tract infections, skin and skin<br />

structure infections: 30mg/kg/day in divided doses every 12 hours. Pharyngitis, tonsillitis:<br />

30mg/kg/day in single or 2 divided doses. For B-hemolytic streptococcal infections, continue<br />

treatment for ≥10 days. Renal function impairment: Adjust dosage according to creatinine<br />

clearance rates to prevent drug accumulation.<br />

Initial adult dose: 1 g; the maintenance dose (based on creatinine clearance rate, ml/min/1.73 m 2 )<br />

is 500mg at the intervals below:<br />

Cefadroxil Dosage in Renal Impairment<br />

Creatinine clearance (ml/min)<br />

Dosage interval (hours)<br />

0-10 36<br />

10-25 24<br />

25-50 12<br />

> 50 No adjustment<br />

Mild infections caused by susceptible gram-positive cocci: 250 to 500 mg every 8 hours.<br />

Moderate-to-severe infections: 500 mg to 1 g every 6 to 8 hours. Pneumococcal pneumonia:<br />

500 mg every 12 hours. Severe, life-threatening infections (e.g., endocarditis, septicemia):<br />

1 to 1.5 g every 6 hours. Rarely, 12 g/day have been used. Acute uncomplicated urinary tract<br />

infections: 1 g every 12 hours.<br />

Perioperative prophylaxis: Preoperative: 1 g IV or IM, 0.5 to 1 hour prior to surgery.<br />

Intraoperative (≥2 hours): 0.5 to 1 g IV or IM during surgery at appropriate intervals.<br />

Postoperative: 0.5 to 1 g IV or IM every 6 to 8 hours for 24 hours after surgery. Prophylactic<br />

administration may be continued for 3 to 5 days, especially where the occurrence of infection may<br />

be particularly devastating (e.g., open heart surgery, prosthetic arthroplasty).<br />

Mild-to-moderately severe infections: A total daily dosage of 25 to 50 mg/kg ( 10 to 20 mg/lb)<br />

in three or four equal doses. Severe infections: Total daily dosage may be increased to 100 mg/kg<br />

(45 mg/lb).<br />

Children: Do not exceed adult recommended doses.<br />

Mild-to-moderately severe infections:<br />

A total daily dosage of 25 to 50 mg/kg ( 10 to 20 mg/lb) in three or four equal doses.<br />

Severe infections:<br />

Total daily dosage may be increased to 100 mg/kg (45 mg/lb).<br />

Renal function impairment:<br />

All reduced dosage recommendations apply after an initial loading dose appropriate to the severity<br />

of the infection.<br />

Cefazolin Dosage in Renal Impairment<br />

Dose<br />

Serum creatinine<br />

(mg/dl)<br />

Ccr<br />

(ml/min)<br />

Mild-tomoderate<br />

infection<br />

(mg)<br />

Moderate-tosevere<br />

infection<br />

(mg)<br />

Dosage<br />

interval<br />

(hrs)<br />

≤1.5 ≥55 250 to 500 500 to 1000 6-8<br />

1.6-3 35-54 250 to 500 500 to 1000 ≥8<br />

3.1-4.5 11-34 125 to 250 250 to 500 12<br />

≥4.6 ≤10 125 to 250 250 to 500 18-24<br />

Adults: 1 to 4g/day in divided doses. Usual dose - 250mg every 6 hours. Streptococcal<br />

pharyngitis, skin and skin structure infections, uncomplicated cystitis in patients > 15 years -<br />

500mg every 12 hours. May need larger doses for more severe infections or less susceptible<br />

organisms. If dose is> 4g/day, use parenteral drugs.<br />

Children: Do not exceed adult recommended doses.<br />

Monohydrate: 25 to 50mg/kg/day in divided doses. For streptococcal pharyngitis in patients > 1<br />

year and for skin and skin structure infections, divide total daily dose and give every 12 hours. In<br />

severe infections, double the dose.<br />

Otitis media: 75 to 100mg/kg/day in 4 divided doses.<br />

B-hemolytic streptococcal infections: Continue treatment for at least 10 days<br />

105


Cephapirin<br />

Powder for<br />

injection:<br />

1gm<br />

Administered by IM or IV injection<br />

• Adults:<br />

o<br />

o<br />

o<br />

500mg – 1 gram every 4-6 hours IM or IV<br />

Serious infections: up to 12grams per day, generally by IV infusion<br />

Alternate dosing: 1-2g q 6 hr for 24 hr.<br />

In clients with impaired renal function, a dose of 7.5-15 mg/kg q 12 hr may<br />

be adequate.<br />

• Perioperative prophylaxis:<br />

o<br />

o<br />

Pre-op: 1-2grams IV or IM, 30 to 60 minutes pre-surgery<br />

Intra-operative: 1-2grams IV or IM during surgery<br />

Cephradine<br />

Cefaclor<br />

Capsules:<br />

250mg, 500mg<br />

Oral suspension:<br />

125mg/5ml<br />

250mg/5ml<br />

Capsules:<br />

250mg, 500mg<br />

Extended-release<br />

tablets:<br />

375mg, 500mg<br />

Oral suspension:<br />

125mg/5ml<br />

187mg/5ml<br />

250mg/5ml<br />

375mg/5ml<br />

o<br />

Post-operative: 1-2grams IV or IM every 6 hours for 24 hours post-op<br />

Children: 40-80mg/kg/day administered in 4 equal doses<br />

Adults:<br />

Skin, skin structures and respiratory tract infections (other than lobar pneumonia):<br />

Usual dose is 250mg every 6 hours or 500mg every 12 hours.<br />

For lobar pneumonia: 500mg every 6 hours or 1 g every 12 hours.<br />

For uncomplicated urinary tract infections: The usual dose is 500mg every 12hours. In more<br />

serious infections and prostatitis, 500mg every 6 hours or 1 g every 12hours. Severe or chronic<br />

infections may require larger doses (≤1g every 6 hours).<br />

Children: No adequate information is available on the efficacy of twice daily regimens in children<br />

< 9 months of age. For children ≥9 months, the usual dose is 25 to 50mg/kg/day, in equally divided<br />

doses every 6 or 12 hours. For otitis media caused by H. influenzae, 75 to 100mg/kg/day in equally<br />

divided doses every 6 or 12 hours is recommended; do not exceed 4 g/day.<br />

All patients, regardless of age and weight: Larger doses (≤1g four times/day) may be given for<br />

severe or chronic infections.<br />

Renal function impairment:<br />

Patients not on dialysis: Use the following initial dosage schedule as a guideline based on<br />

creatinine clearance. Further modification in the dosage schedule may be required because of<br />

individual variations in absorption.<br />

Cephradine Dosage in Renal Impairment<br />

Ccr (ml/min) Dose (mg) Time Interval (hours)<br />

> 20 500 6<br />

5 to 20 250 6<br />

< 5 250 12<br />

Patients on chronic, intermittent hemodialysis: 250 mg initially; repeat at 12 hours and after 36<br />

to 48 hours. Children may require dosage modification proportional to their weight and severity of<br />

infection.<br />

Adults: Usual dosage is 250mg every 8 hours. In severe infections or those caused by less<br />

susceptible organisms, dosage may be doubled. Acute bacterial exacerbations of chronic<br />

bronchitis: 500mg every 12 hours for 7 days. The effectiveness of the extended release tablets<br />

against B-lactamase producing H. influenzae has not been established. Secondary bacterial<br />

infection of acute bronchitis: 500mg every 12 hours for 7 days. The effectiveness of the<br />

extended release tablets against B-lactamase producing H. influenzae has not been established.<br />

Pharyngitis or tonsillitis: 375mg every 12 hours for 10 days. Uncomplicated skin and skin<br />

structure infections: 375mg every 12 hours for 7 to 10 days. The effectiveness of the extended<br />

release tablets against S. pyogenes has not been established.<br />

Children: Give 20mg/kg/day in divided doses every 8 hours. In more serious infections, such as<br />

otitis media and infections caused by less susceptible organisms, administer 40mg/kg/day, with a<br />

maximum dosage of 1g/day. Do not exceed adult recommended doses.<br />

Twice daily treatment option:<br />

For otitis media and pharyngitis, the total daily dosage may be divided and administered every 12<br />

hours. Storage / Stability: Refrigerate suspension after reconstitution; discard after 14 days.<br />

106


Cefprozil<br />

Tablets:<br />

250mg, 500mg<br />

Oral suspension:<br />

125mg/5ml<br />

250mg/5ml<br />

Cefprozil Dosage and Duration<br />

Population/Infection Dosage (mg) Duration (days)<br />

Adults (≥13 years of age)<br />

Pharyngitis/Tonsillitis 500q 24 hrs 10 1<br />

Acute sinusitis (use higher dose for<br />

moderate-to-severe infections)<br />

Secondary bacterial infection of<br />

acute bronchitis and acute<br />

bacterial exacerbation of<br />

chronic bronchitis<br />

Uncomplicated skin and skin<br />

structure infections<br />

Children (2 to 12 years) 2<br />

250q 12 hrs or<br />

500q 12 hrs<br />

10<br />

500q 12 hrs 10<br />

250q 12 hrs,<br />

500q 24 hrs<br />

or 500q 12 hrs<br />

Pharyngitis/Tonsillitis 7.5 mg/kg q12 hrs 10 1<br />

Uncomplicated skin and skin<br />

structure infections<br />

Infants and children(6 months to<br />

12 years) 2<br />

10<br />

20 mg/kg q24 hrs 10<br />

Otitis media 15 mg/kg q12 hrs 10<br />

Acute sinusitis (use higher dose<br />

7.5 mg/kg q12 hrs or<br />

for moderate-to-severe infections)<br />

15 mg/kg q12 hrs<br />

1 For infections caused by S. pyogenes, administer for 10 days.<br />

2 Not to exceed adult recommended doses.<br />

10<br />

Cefuroxime<br />

Tablets:<br />

125mg, 250mg,<br />

500mg<br />

Oral suspension:<br />

125mg/5ml<br />

250mg/5ml<br />

Powder for<br />

injection:<br />

750mg<br />

Renal function impairment:<br />

For creatinine clearance (Ccr) of 30 to 120ml/min, use standard dosage and dosing interval. For<br />

Ccr < 30 ml/min, use a dosage 50% of standard at the standard dosing interval.<br />

Oral : Tablets and suspension are NOT bioequivalent and are NOT substitutable on a mg/mg<br />

basis. Tablets: The tablets may be given without regard to meals.<br />

Dosage for Cefuroxime Axetil Tablets<br />

Adults (≥13 years)<br />

Population/Infection Dosage Duration (days)<br />

Pharyngitis/tonsillitis 250mg bid 10<br />

Acute bacterial exacerbations of chronic bronchitis 1 10<br />

Secondary bacterial infections of acute<br />

250 or 500mg bid 5-10<br />

bronchitis<br />

Uncomplicated skin and skin structure<br />

infections<br />

250 or 500mg bid 10<br />

Uncomplicated urinary tract infections 125 or 250mg bid 7 to 10<br />

Uncomplicated gonorrhea 1000mg once single dose<br />

Early Lyme disease 500mg bid 20<br />

Children who can swallow tablets whole 2<br />

Pharyngitis/tonsillitis 125mg bid 10<br />

Acute otitis media 250mg bid 10<br />

1 Safety and efficacy of drug administered < 10 days in patients with acute exacerbations of chronic<br />

bronchitis have not been established.<br />

2 Do not exceed adult recommended doses.<br />

Suspension: Must be administered with food. Shake well each time before using. The suspension<br />

may be administered to children ranging in age from 3 months to 12 years, according to dosages in<br />

the following table:<br />

Dosage for Cefuroxime Axetil Suspension<br />

Infection<br />

(infants and children,<br />

3 months to 12 years ) Dosage<br />

Daily maximum<br />

dose<br />

Duration<br />

(days)<br />

Pharyngitis/tonsillitis 20mg/kg/day divided bid 500 mg 10<br />

Acute otitis media 30mg/kg/day divided bid 1000 mg 10<br />

Impetigo 30mg/kg/day divided bid 1000 mg 10<br />

Renal function impairment: Because cefuroxime is renally eliminated, its half-life will be<br />

107


Cefdinir<br />

Capsule: 300mg<br />

Oral suspension:<br />

125mg/5ml<br />

prolonged in patients with renal failure.<br />

Adults/Adolescents: The recommended dosage and duration of treatment for infections in adults<br />

and adolescents are described in the following chart; the total daily dose for all infections is<br />

600mg. Once-daily dosing for 10 days is as effective as twice-daily dosing. Once-daily dosing has<br />

not been studied in pneumonia or skin infections; therefore, administer twice daily in these<br />

infections. Capsules may be taken without regard to meals.<br />

Cefdinir Dosage in Adults and Adolescents (≥13 years of age)<br />

Type of infection Dosage Duration<br />

Community-acquired pneumonia 300mg q 12 hrs 10 days<br />

Acute exacerbations of chronic bronchitis<br />

Acute maxillary sinusitis<br />

Pharyngitis/Tonsillitis<br />

300mg q 12 hrs<br />

or<br />

600mg q 24 hrs<br />

300mg q 12 hrs<br />

or<br />

600mg q 24 hrs<br />

300mg q 12 hrs<br />

or<br />

600mg q 24 hrs<br />

10 days<br />

10 days<br />

10 days<br />

10 days<br />

5 to 10 days<br />

10 days<br />

Uncomplicated skin and skin structure<br />

300mg q 12 hrs 10 days<br />

infections<br />

Children (6 months through 12 years of age):<br />

The recommended dosage and duration of treatment for infections in pediatric patients are<br />

described in the following chart; the total daily dose for all infections is 14mg/kg, up to a<br />

maximum dose of 600mg/day. Once-daily dosing for 10 days is as effective as twice-daily dosing.<br />

Once-daily dosing has not been studied in skin infections; therefore, administer twice daily in this<br />

infection. Oral suspension may be administered without regard to meals.<br />

Cefdinir Dosage in Pediatric Patients (Age 6 Months Through 12 years)<br />

Acute bacterial otitis media<br />

Acute maxillary sinusitis<br />

Pharyngitis/Tonsillitis<br />

Type of infection Dosage Duration<br />

Uncomplicated skin and skin structure<br />

infections<br />

7 mg/kg q 12 h<br />

or<br />

14 mg/kg q 24 h<br />

7 mg/kg q 12 h<br />

or<br />

14 mg/kg q 24 h<br />

7 mg/kg q 12 h<br />

or<br />

14 mg/kg q 24 h<br />

10 days<br />

10 days<br />

10 days<br />

10 days<br />

5 to 10 days<br />

10 days<br />

7 mg/kg q 12 h 10 days<br />

kg<br />

Cefdinir for Oral Suspension Pediatric Dosage Chart<br />

Weight<br />

lb<br />

125 mg/5 ml<br />

9 20 2.5 ml (½ tsp) q 12 h<br />

or 5 ml (1 tsp) q 24 h<br />

18 40 5 ml (1 tsp) q 12 h<br />

or 10 ml (2 tsp) q 24 h<br />

27 60 7.5 ml (1½ tsp) q 12 h<br />

or 15 ml (3 tsp) q 24 h<br />

36 80 10 ml (2 tsp) q 12 h<br />

or 20 ml (4 tsp) q 24 h<br />

≥43 1 95 12 ml (2½ tsp) q 12 h<br />

or 24 ml (5 tsp) q 24 h<br />

1 Pediatric patients who weigh ≥43 kg should receive the maximum daily dose of 600 mg.<br />

Renal function impairment:<br />

For adult patients with creatinine clearance < 30 ml/min, the dose of cefdinir should be 300 mg<br />

given once daily. Creatinine clearance is difficult to measure in outpatients. However, the<br />

following formula may be used to estimate creatinine clearance (Ccr) in adult patients. For<br />

estimates to be valid, serum creatinine levels should reflect steady-state levels of renal function.<br />

108


Cefditoren Tablets: 200mg Cefditoren Dosage and Administration<br />

in Adults and Adolescents ≥12 Years of Age 1<br />

Type of infection Dosage Duration (days)<br />

Acute bacterial exacerbation 400mg BID<br />

of chronic bronchitis<br />

Pharyngitis/Tonsillitis<br />

10<br />

Uncomplicated skin and skin 200mg BID<br />

structure infections<br />

1 Take with meals.<br />

Renal impairment: No dose adjustment is necessary for patients with mild renal impairment(Ccr<br />

50 to 80mL/min/1.73 m 2 ). It is recommended that ≤200mg twice daily be administered to patients<br />

with moderate renal impairment (Ccr 30 to 49 mL/min/1.73 m 2 ) and 200mg every day be<br />

administered to patients with severe renal impairment (Ccr < 30mL/min/1.73 m 2 ). The appropriate<br />

dose in patients with end-stage renal disease has not been determined.<br />

Cefepime<br />

Powder for<br />

injection:<br />

500mg, 1gm,<br />

2gm<br />

Recommended Dosage Schedule for Cefepime<br />

Site and type of infection Dose Frequency<br />

Duration<br />

(days)<br />

Mild-to-moderate uncomplicated or complicated 0.5 to 1g IV/IM 2 Q12 hrs 7 to 10<br />

E. coli, K. pneumoniae, or P. mirabilis. 1<br />

urinary tract infections, including pyelonephritis,<br />

caused by<br />

Severe uncomplicated or complicated urinary tract<br />

2g IV Q12 hrs 10<br />

or K. pneumoniae. 1<br />

infections, including pyelonephritis, caused by E. coli<br />

Moderate-to-severe pneumonia caused by S.<br />

1 to 2g IV Q12 hrs 10<br />

pneumoniae, 1 Pseudomonas aeruginosa, Klebsiella<br />

pneumoniae, or Enterobacter sp.<br />

Moderate-to-severe uncomplicated skin and skin<br />

2g IV Q12 hrs 10<br />

structure infections caused by S. aureus or S. pyogenes.<br />

Empiric therapy for febrile neutropenic patients. 2g IV Q8 hrs 7 3<br />

Complicated intra-abdominal infections (used in<br />

2g IV q12 hrs 7 to 10<br />

combination with metronidazole) caused by E. coli,<br />

viridans group streptococci, P. aeruginosa, K.<br />

pneumoniae, Enterobacter species, or B. fragilis.<br />

1 Including cases associated with concurrent bacteremia.<br />

2 IM route of administration is indicated only for mild-to-moderate, uncomplicated, or complicated UTIs<br />

caused by E. coli when the IM route is a more appropriate route of drug administration.<br />

3 Or until resolution of neutropenia. In patients whose fever resolves but who remain neutropenic for > 7<br />

days,<br />

the need for continued antimicrobial therapy should be reevaluated frequently.<br />

Renal function impairment:<br />

In patients with impaired renal function (creatinine clearance ≤60 ml/min), adjust the dose of cefepime to<br />

compensate for the slower rate of renal elimination. The recommended initial dose should be the same as<br />

in patients with normal renal function. In patients undergoing hemodialysis, 68% of the total amount<br />

of<br />

Cefepime present in the body at the start of dialysis will be removed during a 3-hour dialysis period. A<br />

repeat dose, equivalent to the initial dose, should be given at the completion of each dialysis session. In<br />

elderly patients, adjust dosage and administration in the presence of renal insufficiency. In patients<br />

undergoing continuous ambulatory peritoneal dialysis, administer cefepime at normal recommended<br />

doses at a dosage interval of every 48 hours.<br />

Recommended Cefepime Maintenance Schedule in Patients<br />

with Renal Impairment<br />

Creatinine<br />

clearance<br />

(ml/min)<br />

> 60<br />

Recommended maintenance schedule<br />

500mg<br />

q 12 hrs 1 1g q 12 hrs 2g q 12 hrs 2g q 8 hrs<br />

30 to 60 500mg 1g q 24 hrs 2g q 24 hrs 2g q 12 hrs<br />

11 to 29 q 24 hrs 500mg q 24 hrs 1g q 24 hrs 2g q 24 hrs<br />

250mg<br />

250mg q 24 hrs 500mg q 24 hrs 1g q 24 hrs<br />

< 11 q 24 hrs<br />

1 Normal recommended dosing schedule.<br />

109


Cefixime<br />

Cefotaxime<br />

Powder for<br />

injection:<br />

500mg, 1gm,<br />

2gm<br />

Powder for<br />

injection:<br />

500mg, 1gm,<br />

2gm, 10gm<br />

Injection:<br />

1gm, 2gm<br />

Recommended Dosage Schedule for Cefepime<br />

Site and type of infection Dose Frequency<br />

Duration<br />

(days)<br />

Mild-to-moderate uncomplicated or complicated<br />

urinary tract infections, including pyelonephritis,<br />

caused by<br />

E. coli, K. pneumoniae, or P. mirabilis. 1 0.5 to 1g IV/IM 2 Q12 hrs 7 to 10<br />

Severe uncomplicated or complicated urinary tract<br />

infections, including pyelonephritis, caused by E.<br />

coli<br />

or K. pneumoniae. 1 2g IV Q12 hrs 10<br />

Moderate-to-severe pneumonia caused by S.<br />

pneumoniae, 1 Pseudomonas aeruginosa, Klebsiella<br />

pneumoniae, or Enterobacter sp.<br />

Moderate-to-severe uncomplicated skin and skin<br />

structure infections caused by S. aureus or S.<br />

pyogenes.<br />

1 to 2g IV Q12 hrs 10<br />

2g IV Q12 hrs 10<br />

Empiric therapy for febrile neutropenic patients. 2g IV Q8 hrs 7 3<br />

Complicated intra-abdominal infections (used in 2g IV Q12 hrs 7 to 10<br />

combination with metronidazole) caused by E.<br />

coli,<br />

viridans group streptococci, P. aeruginosa, K.<br />

pneumoniae, Enterobacter species, or B. fragilis.<br />

1 Including cases associated with concurrent bacteremia.<br />

2 IM route of administration is indicated only for mild-to-moderate, uncomplicated, or complicated UTIs<br />

caused by E. coli when the IM route is a more appropriate route of drug administration.<br />

3 Or until resolution of neutropenia. In patients whose fever resolves but who remain neutropenic for > 7<br />

days, the need for continued antimicrobial therapy should be reevaluated frequently.<br />

Renal function impairment: In patients with impaired renal function (creatinine clearance ≤60<br />

ml/min),<br />

adjust the dose of cefepime to compensate for the slower rate of renal elimination. The recommended<br />

initial dose should be the same as in patients with normal renal function.<br />

In patients undergoing hemodialysis, 68% of the total amount of cefepime present in the body at the<br />

start<br />

of dialysis will be removed during a 3-hour dialysis period. A repeat dose, equivalent to the initial dose,<br />

should be given at the completion of each dialysis session. In elderly patients, adjust dosage and<br />

administration in the presence of renal insufficiency. In patients undergoing continuous ambulatory<br />

peritoneal dialysis, administer cefepime at normal recommended doses at a dosage interval of every 48<br />

hours.<br />

Recommended Cefepime Maintenance Schedule in Patients<br />

with Renal Impairment<br />

Creatinine<br />

clearance<br />

(ml/min)<br />

> 60<br />

Recommended maintenance schedule<br />

500mg<br />

q 12 hrs 1 1g q 12 hrs 2g q 12 hrs 2g q 8 hrs<br />

30 to 60 500mg 1g q 24 hrs 2g q 24 hrs 2g q 12 hrs<br />

11 to 29 q 24 hrs 500mg q 24 hrs 1g q 24 hrs 2g q 24 hrs<br />

250mg<br />

< 11 q 24 hrs<br />

250mg q 24 hrs 500mg q 24 hrs 1g q 24 hrs<br />

1 Normal recommended dosing schedule.<br />

Adults: Administer IV or IM. The maximum daily dosage should not exceed 12 g. Determine dosage<br />

and route of administration by susceptibility of the causative organisms, severity of the infection and the<br />

patient's condition (see table for dosage guidelines).<br />

Cefotaxime Dosage Guidelines for Adults<br />

Daily<br />

Type of infection<br />

dosage (g) Frequency and route<br />

Gonococcal urethritis/cervicitis in males<br />

and females 0.5 0.5g IM (single dose)<br />

Rectal gonorrhea in females 0.5 0.5g IM (single dose)<br />

110


Cefpodoxime<br />

Ceftazidime<br />

Tablets:<br />

100mg, 200mg<br />

Granules for<br />

Suspension<br />

50mg/5ml<br />

100mg/5ml<br />

Powder for<br />

injection:<br />

500mg, 1gm<br />

2gm, 6gm<br />

Injection:<br />

1gm<br />

Rectal gonorrhea in males 1 1g IM (single dose)<br />

Uncomplicated infections 2 1g every 12 hours<br />

IM or IV<br />

Moderate-to-severe 3 to 6 1 to 2g every 8 hours<br />

IM or IV<br />

Infections commonly needing<br />

higher dosage (e.g., septicemia)<br />

6 to 8 2g every 6 to<br />

8 hours IV<br />

Life-threatening infections 12 2g every 4 hours IV<br />

Perioperative prophylaxis: 1 g IV or IM, 30 to 90 minutes prior to surgery.<br />

Cesarean section: Administer the first 1 g dose IV as soon as the umbilical cord is clamped.<br />

Administer the second and third doses as 1 g IV or IM at 6 and 12 hour intervals after the first dose.<br />

Pediatric: It is not necessary to differentiate between premature and normal gestational age infants.<br />

The following dosage recommendations may serve as a guide:<br />

Cefotaxime Dosage Guidelines in Pediatric Patients<br />

Age Weight (kg) Dosage schedule Route<br />

0 to 1 week -- 50 mg/kg every 12 hours IV<br />

1 to 4 weeks -- 50 mg/kg every 8 hours IV<br />

1 month to 12 years < 50 1 50 to 180 mg/kg/day IV or IM<br />

in 4 to 6 divided doses 2<br />

1 For children ≥50 kg, use adult dosage. Do not exceed adult recommended doses.<br />

2 Use higher doses for more severe or serious infections including meningitis.<br />

Renal function impairment: Determine dosage by degree of renal impairment, severity of<br />

infection and susceptibility of the causative organism. In patients with estimated creatinine clearances<br />

of < 20 ml/min/1.73 m 2 , reduce dosage by 50%.<br />

Administer tablets with food to enhance absorption. Administer oral suspension without regard to food.<br />

Dosage/Duration of Cefpodoxime<br />

Total daily<br />

Type of infection<br />

dose<br />

Adults ≥13 years of age<br />

Dose<br />

frequency<br />

Duration<br />

Acute community-acquired pneumonia 400mg 200mg every 12 hrs 14 days<br />

Acute bacterial exacerbations of chronic bronchitis<br />

(tablets) 400mg 200mg every 12 hrs 10 days<br />

Uncomplicated gonorrhea(men and women) and<br />

rectal gonococcal infections (women) 200mg single dose<br />

Skin and skin structure<br />

Pharyngitis/tonsillitis<br />

800mg<br />

200mg<br />

400mg every 12 hrs<br />

100mg every 12 hrs<br />

7 to 14<br />

days<br />

5 to 10<br />

days<br />

Uncomplicated urinary tract infection 200mg 100mg every 12 hrs 7 days<br />

Children(age 5 months through 12 years): 1<br />

Acute otitis media<br />

Pharyngitis/tonsillitis<br />

10mg/kg/day<br />

(max 400<br />

mg/day)<br />

10mg/kg/day<br />

(max 200<br />

mg/day)<br />

10mg/kg every 24 hrs<br />

(max 400mg/dose)or<br />

5mg/kg every 12 hrs<br />

(max 200mg/dose)<br />

5mg/kg every 12hours<br />

(max 100 mg/dose)<br />

10 days<br />

5 to 10<br />

days<br />

1 Do not exceed adult recommended doses.<br />

Renal function impairment: For patients with severe renal impairment (creatinine clearance [Ccr]


Ceftibuten<br />

Capsule: 400mg<br />

Powder for oral<br />

suspension:<br />

90mg/5ml<br />

Uncomplicated pneumonia; mild skin<br />

and skin structure infections<br />

500mg to 1g IV or IM<br />

Q8 hrs<br />

Bone and joint infections 2g IV Q12 hrs<br />

Serious gynecological and intraabdominal<br />

infections<br />

Meningitis<br />

Very severe life-threatening infections,<br />

especially in immunocompromised<br />

patients<br />

Pseudomonal lung infections in<br />

cystic fibrosis patients w/normal<br />

renal function 1<br />

2g IV<br />

30 to 50mg/kg IV<br />

to a max 6g/day<br />

Q8 hrs<br />

Q8 hrs<br />

Neonates (0 to 4 weeks) 2 30mg/kg IV Q12 hrs<br />

Infants and children<br />

30 to 50mg/kg IV Q8 hrs<br />

(1 month to 12 years) 2 to 6g/day 3<br />

1 Although clinical improvement has been shown, bacteriological cures cannot be expected in patients<br />

with chronic respiratory disease and cystic fibrosis.<br />

2 Do not exceed adult recommended doses.<br />

3 Reserve the higher dose for immunocompromised children or children with cystic fibrosis or meningitis.<br />

Hepatic function impairment: No dosage adjustment is required.<br />

Renal function impairment: Ceftazidime is excreted by the kidneys, almost exclusively by glomerular<br />

filtration. In patients with impaired renal function (GFR < 50 ml/min), reduce dosage to compensate<br />

for slower excretion. In patients with suspected renal insufficiency, give an initial loading dose of 1 g.<br />

Estimate GFR to determine the appropriate maintenance dose.<br />

Ceftibuten suspension must be administered ≥2 hours before or 1 hour after a meal.<br />

Ceftibuten Dosage and Duration<br />

Type of infection<br />

Adults ≥12 years of age<br />

Acute bacterial exacerbations of<br />

chronic bronchitis caused by<br />

H. influenzae, M. catarrhalis<br />

or Streptococcus pneumoniae<br />

Pharyngitis and tonsillitis caused by<br />

S. pyogenes<br />

Acute bacterial otitis media caused<br />

by H. influenzae, M. catarrhalis<br />

or S. pyogenes<br />

Children 1<br />

Pharyngitis and tonsillitis caused by S. pyogenes<br />

Acute bacterial otitis media caused<br />

by H. influenzae, M. catarrhalis<br />

or S. pyogenes<br />

1 Do not exceed adult recommended doses.<br />

Daily maximum<br />

dose<br />

Dose and<br />

frequency<br />

Duration<br />

400mg 400mg QD 10 days<br />

400mg 9mg/kg QD 10 days<br />

Ceftizoxime<br />

Powder for<br />

injection:<br />

500mg, 1gm<br />

2gm, 10gm<br />

Injection:<br />

1gm, 2gm<br />

Ceftibuten Oral Suspension<br />

Pediatric Dosage Chart 1<br />

Weight<br />

kg lb 90 mg/5 ml 180 mg/5 ml<br />

10 22 5 ml (1 tsp) QD 2.5 ml (1/2 tsp) QD<br />

20 44 10 ml (2 tsp) QD 5 ml (1 tsp) QD<br />

40 88 20 ml (4 tsp) QD 10 ml (2 tsp) QD<br />

1 Children > 45 kg should receive the maximum daily dose of 400 mg<br />

Adults: Usual dosage is 1 or 2 g every 8 to 12 hours. Individualize dosage.<br />

Ceftizoxime Dosage Guidelines in Adults<br />

Type of infection Daily dose (g) Frequency and route<br />

Uncomplicated urinary tract 1 500mg every 12 hours IM or IV<br />

PID 1 6 2g every 8 hours IV<br />

Other sites 2-3 1g every 8 to 12 hours IM or IV<br />

Severe or refractory 3-6 1g every 8 hours IM or IV<br />

112


Life-threatening 2 9-12 3 to 4g every 8 hours IV<br />

1 Dosages ≤2 g every 4 hours have been given.<br />

2 Divide 2 g IM doses and give in different large muscle masses.<br />

Urinary tract infections: Because of the serious nature of urinary tract infections caused<br />

by P. aeruginosa and because many strains of Pseudomonas species are only moderately<br />

susceptible to ceftizoxime, higher dosage is recommended. Institute other therapy if the response<br />

is not prompt.<br />

Gonorrhea, uncomplicated:<br />

A single 1g IM injection is the usual dose.<br />

Life-threatening infections:<br />

The IV route may be preferable for patients with bacterial septicemia, localized parenchymal abscesses<br />

(such as intra-abdominal abscess), peritonitis or other severe or life-threatening infections. In those<br />

patients with normal renal function, the IV dosage is 2 to 12 g/day. In conditions such as bacterial<br />

septicemia, 6 to 12 g/day IV may be given initially for several days, and the dosage gradually reduced<br />

according to clinical response and laboratory findings.<br />

Pediatric: Children (6 months): 50mg/kg every 6 to 8 hours. Dosage may be increased to<br />

200mg/kg/day.<br />

Do not exceed the maximum adult dose for serious infection.<br />

Renal function impairment: Requires modification of dosage. Following an initial loading dose of<br />

500mg<br />

to 1g IM or IV, use the maintenance dosing schedule in the following table. Determine further dosing by<br />

therapeutic monitoring, severity of the infection and susceptibility of the causative organisms.<br />

Ceftriaxone<br />

Powder for<br />

injection:<br />

250mg, 500mg,<br />

1gm<br />

2gm, 10gm<br />

Injection:<br />

1gm, 2gm<br />

Administer IV or IM. Continue for ≥2 days after signs and symptoms of infection have disappeared.<br />

Usual duration is 4 to 14 days; in complicated infections, longer therapy may be required. For<br />

S. pyogenes, continue for ≥10 days.<br />

Adults: Usual daily dose is 1 to 2g once a day (or in equally divided doses twice a day) depending<br />

on the type and severity of the infection. Do not exceed total daily dose of 4g.<br />

Uncomplicated gonococcal infections: Give a single IM dose of 250mg.<br />

Surgical prophylaxis: Give a single 1g dose IV 0.5 to 2 hours before surgery.<br />

Children: To treat serious infections other than meningitis, administer 50 to 75mg/kg/day<br />

(not to exceed 2 g) in divided doses every 12 hours.<br />

Meningitis: 100mg/kg/day (not to exceed 4g). Thereafter, a total daily dose of 100mg/kg/day<br />

(not to exceed 4g/day) is recommended. May give daily dose once per day or in equally divided<br />

doses every 12 hours. Usual duration is 7 to 14 days.<br />

Skin and skin structure infections: Give 50 to 75mg/kg once daily (or in equally divided doses twice<br />

daily),<br />

not to exceed 2g.<br />

Renal/hepatic function impairment: No dosage adjustment is necessary; however, monitor blood levels.<br />

In patients with both hepatic dysfunction and significant renal disease, the dose should not exceed 2g/day<br />

without closely monitoring serum concentrations.<br />

CDC recommended treatment schedules for chancroid, gonorrhea and acute pelvic inflammatory<br />

disease (PID)<br />

Chancroid (Haemophilus ducreyi infection): 250mg IM as a single dose.<br />

Gonococcal infections:<br />

Uncomplicated: 125mg IM in a single dose plus 1g azithromycin in single oral dose or 100mg<br />

doxycycline twice a day for 7 days.<br />

Conjunctivitis: 1g IM single dose.<br />

Disseminated: 1g IM or IV every 24 hours.<br />

Meningitis/Endocarditis: 1 to 2g IV every 12 hours for 10 to 14 days (meningitis) or for 4 weeks<br />

(endocarditis).<br />

Special Dosing Considerations:<br />

The pregnancy category is Category “B” for the cephalosporins. 7 Safety during pregnancy is not<br />

established. Use only when potential benefits outweigh potential hazards to the fetus.<br />

All cephalosporin require dosing adjustment in renal impaired patients. Reduce total daily dose in patients<br />

with transient or persistent reduction of urinary output caused by renal insufficiency, high and prolonged<br />

serum concentrations can occur in such patients from usual doses.<br />

113


Table 7. Special Dosing Considerations for the Cephalosporins<br />

Drug Pediatric Use Special consideration<br />

Cefadroxil Maximum dose: 2 grams/day Available in tablet, capsule, and oral suspension for<br />

flexible dosing/route of administration.<br />

Cefazolin Safety and efficacy of capsule and suspension in<br />

Parenteral only.<br />

children<br />


divided doses every 12 hours.<br />

pain of IM injections. Parenteral only.<br />

115


VIII. Comparative Effectiveness of the Cephalosporins<br />

Table 8 describes comparative clinical data for the cephalosporins.<br />

Table 8. Outcomes Evidence for the Cephalosporins<br />

Reference Study Design Entry Criteria N Treatment Regimen Duration<br />

of Study<br />

Upper Respiratory Infection<br />

Pessey at<br />

al. 8<br />

Randomized,<br />

open,<br />

multicenter<br />

• Aged 6-36 months<br />

with acute otitis media<br />

with effusion,<br />

diagnosed by<br />

tympanocentesis and<br />

microbiologic culture<br />

• No antimicrobial<br />

treatment in the past 72<br />

hours<br />

716 • Cefuroxime axetil<br />

30mg/kg/day in two<br />

divided doses for 5 days.<br />

• Amoxicillin/<br />

clavulanate 40mg/kg/day<br />

in three divided doses for<br />

8 days<br />

• Amoxicillin/<br />

clavulanate 40mg/kg/day<br />

in three divided doses for<br />

10 days<br />

TOC =<br />

Day 8<br />

LTFU =<br />

Day 28<br />

Results<br />

Primary Endpoint:<br />

• Clinical response at the posttreatment assessment. Patients<br />

were considered cured if the clinical signs and symptoms<br />

subsided within the treatment period (exclusive of middle ear<br />

effusion) and were absent at posttreatment and follow-up;<br />

bacteriologic cure was presumed in these patients. Failure was<br />

defined as lack of resolution of clinical signs and symptoms<br />

(exclusive of middle ear effusion) and relapse as the<br />

reappearance, after initial cure, of clinical signs or symptoms<br />

of the original infection up to 28 days posttreatment.<br />

• Safety was assessed from all reported adverse events.<br />

Efficacy: cefuroxime 5 days = amoxicillin/clavulanate 8 days =<br />

amoxicillin/clavulanate 10 days<br />

• Primary endpoint was measured as clinical signs and<br />

symptoms subsided within the treatment period. Patients were<br />

evaluated at day 1, 8, and 28.<br />

• 86% in cefuroxime 5 day group, 88% in<br />

amoxicillin/clavulanate 8 day group and 88% in<br />

amoxicillin/clavulanate 10 day group responded to the therapy.<br />

• There was no difference in efficacy measurement among the 3<br />

study groups.<br />

Safety: cefuroxime 5 days = amoxicillin/clavulanate 8 days ><br />

amoxicillin/clavulanate 10 days<br />

• The incidence of diarrhea was higher in<br />

amoxicillin/clavulanate 10 day group than in<br />

amoxicillin/clavulanate 8 day group and the cefuroxime group.<br />

Nemeth et Randomized, • Patients ≥ 13 years 919 • Cefdinir 600mg daily TOC = 4-9 Primary Endpoint:


al. 9<br />

Pichichero<br />

et al. 10<br />

open,<br />

double-mask,<br />

multicenter<br />

Randomized,<br />

observer-blind,<br />

multicenter<br />

of age with diagnosis<br />

of group A streptococci<br />

pharyngitis<br />

• Children aged 2 to<br />

17 years with signs and<br />

symptoms of acute<br />

tonsillopharyngitis<br />

• Children with<br />

positive throat culture<br />

for group A<br />

streptococcus were<br />

evaluated<br />

487<br />

(377<br />

were<br />

evalu<br />

ated)<br />

for 10 days<br />

• Cefdinir 300mg twice<br />

daily for 10 days<br />

• Penicillin V 250mg<br />

four times daily for 10<br />

days<br />

• Cefpodoxime 5mg/kg<br />

twice daily for 5 days<br />

• Cefpodoxime<br />

10mg/kg per day for 10<br />

days<br />

• Penicillin V 40mg/kg<br />

per day divided into 3<br />

doses for 10 days<br />

days<br />

LTFU =<br />

17-24 days<br />

Post<br />

therapy<br />

TOC =<br />

Day 3-17<br />

LTFU =<br />

Day 32-38<br />

Asmar et Randomized, • Patients aged 2 368 • Cefpodoxime TOC = Primary Endpoints:<br />

• Clinically cured – All signs and symptoms are absent or in<br />

satisfactory remission and no further antibacterial therapy is<br />

required at long term follow-up<br />

• Microbiologic eradication – Cultures are negative for group-<br />

A beta hemolytic streptococci<br />

Efficacy: cefdinir 600mg QD=cefdinir 300mg BID>penicillin<br />

• The clinical cure rates at the test of cure were 94.8% for the<br />

cefdinir 600mg QD group, 96.3% for the cefdinir 300mg BID<br />

group and 88.9% for the penicillin group. The clinical cure rates<br />

in both cefdinir groups were significantly better than the<br />

penicillin group.<br />

• The microbiology eradication rates were 91.4% for the<br />

cefdinir 600mg QD group, 91.7% for the cefdinir 300mg BID<br />

group and 83.4% for the penicillin group. Both cefdinir groups<br />

were superior to the penicillin group.<br />

Safety: both cefdinir groups < penicillin<br />

• Diarrhea was more common in the cefdinir groups. (P cefpodoxime 5 day≥ penicillin<br />

• The clinical response rate was higher in cefpodoxime 10 day<br />

group (96%) than the cefpodoxime 5 day group (94%) and<br />

penicillin group (91%). The results did not reach statistical<br />

significance.<br />

• The bacteriologic eradication rate for cefpodoxime 10 day<br />

group was 95%, cefpodoxime 5 day group was 90% and<br />

penicillin 78%.<br />

Safety: cefpodoxime 10 day= cefpodoxime 5 day=penicillin<br />

• There was no difference in adverse events between treatment<br />

groups.<br />

• The most common adverse event was GI distress.<br />

117


al. 11<br />

multicenter,<br />

investigatorblind<br />

Lower Respiratory Infection<br />

Fogarty et Prospective,<br />

al. 12 randomized,<br />

double-blind,<br />

multicenter,<br />

Parallel<br />

Paster, et<br />

al. 13<br />

Randomized,<br />

double-blind<br />

multicenter<br />

months to 17 years<br />

with a diagnosis of<br />

acute suppurative otitis<br />

media.<br />

• Chronic bronchitis<br />

defined as cough and<br />

sputum production for<br />

at least three<br />

consecutive months<br />

• Age ≥ 13 years<br />

• Absence of an<br />

infiltrate on chest X ray<br />

• Patients were at least<br />

13 years old with<br />

diagnosis of acute<br />

exacerbation of chronic<br />

(236<br />

were<br />

evaluated)<br />

10mg/kg once daily for<br />

10 days<br />

• Cefixime 8mg/kg<br />

once daily for 10 days<br />

548 • Cefprozil 500mg<br />

twice daily (median<br />

time 10 days)<br />

• Cefdinir 300mg<br />

twice daily (median time<br />

5 days)<br />

586 • Cefdinir 300mg twice<br />

daily for 5 days<br />

• Loracarbef 400mg<br />

twice daily for 7 days<br />

Day 12-15<br />

LTFU =<br />

Day 25-38<br />

Cefdinir<br />

pts were<br />

evaluated<br />

on days 12-<br />

16;<br />

Cefprozil<br />

pts were<br />

evaluated<br />

on days 17-<br />

21<br />

TOC =<br />

Day 12-19<br />

LTFU =<br />

Days 26-40<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

Efficacy: cefpodoxime = cefixime<br />

• The clinical response rates were 56% for cefpodoxime treated<br />

patients and 54% for cefixime patients.<br />

• At long-term follow-up, 17% of the patients in cefpodoxime<br />

group and 20% in cefixime group had a recurrent infection.<br />

Safety: cefpodoxime = cefixime<br />

• Drug related adverse events (diarrhea, diaper rash, vomiting)<br />

occurred in 23.3% of cefpodoxime-treated patients and 17.9% of<br />

cefixime-treated patients. (P=0.282).<br />

Primary Endpoints:<br />

• Overall clinical efficacy - Clinical cure was defined as<br />

absence or significant remission of all admission signs and<br />

symptoms. Failure was defined as lack of significant remission<br />

of signs and symptoms and/or the requirement for further<br />

antibiotic treatment. Recurrence was defined as worsening of<br />

signs and symptoms at the LTFU after cure at the TOC visit.<br />

• Bacteriologic eradication rate<br />

• Occurrence of adverse events<br />

Efficacy: cefprozil = cefdinir<br />

• The clinical cure rate was similar in both groups: 80% cefdinir<br />

and 72% cefprozil, 95% CIs were between –1.6% and 18.3%.<br />

• The microbiological eradication of pathogen rates was 81%<br />

for cefdinir-treated patients and 84% for cefprozil-treated<br />

patients.<br />

Safety: cefprozil ≥ cefdinir<br />

• 34% cefdinir treated patients and 33% cefprozil treated<br />

patients experienced at least one adverse event.<br />

• 17% cefdinir patients versus 6% cefprozil patients reported<br />

diarrhea. (P


Drehobl, et<br />

al. 14<br />

Phillips, et<br />

al. 15<br />

Randomized,<br />

double-blind,<br />

multicenter<br />

Randomized,<br />

multicenter,<br />

observer-blind<br />

bronchitis Efficacy: cefdinir 300mg for 5 days = loracarbef 400mg for 7<br />

days<br />

• Clinical response was determined by absence of clinical signs<br />

and symptoms at days 12-19 and days 26-40.<br />

• The response rates were 86% in cefdinir group and 85% in<br />

loracarbef group.<br />

• The microbiology eradication rates were 88% for cefdinir<br />

group and 90% in loracarbef group.<br />

• Predominant pathogens were H. parainfluenzae, H. influenzae,<br />

M. Catarrhalis and S. aureus.<br />

Safety: cefdinir > loracarbef<br />

• Adverse event rates were 30% for cefdinir and 21%<br />

loracarbef.<br />

• The most common adverse events were diarrhea for both<br />

groups, nausea for cefdinir and headache for loracarbef.<br />

• Patients ≥ 13 years<br />

of age with a diagnosis<br />

of community-acquired<br />

pneumonia<br />

• Patients who<br />

received antibiotics<br />

within 7 days of<br />

enrollment were<br />

excluded.<br />

• Patients with signs or<br />

symptoms of acute<br />

bacterial exacerbation<br />

of COPD<br />

690 • Cefdinir 300mg twice<br />

daily for 10 days<br />

• Cefaclor 500mg three<br />

times day for 10 days<br />

301 • Cefpodoxime 200mg<br />

twice daily for 10 days<br />

• Cefaclor 250mg three<br />

times a day for 10 days<br />

TOC =<br />

6-14 days<br />

LTFU =<br />

21-35 days<br />

posttherapy<br />

TOC =<br />

Day 3-7<br />

posttherapy<br />

LTFU =<br />

4 weeks<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefdinir = cefaclor<br />

• The clinical response (cure plus improvement) rates were 89%<br />

in the cefdinir group and 86% in the cefaclor group.<br />

• 78% of patients with isolated pathogens were evaluated for<br />

microbiologic eradication. The rates were 92% in the cefdinir<br />

group and 93% in cefaclor group.<br />

• The predominant pathogens were H. parainfluenzae, H.<br />

influenza, S. pneumonia and S. aureus.<br />

Safety: cefdinir ≥ cefaclor<br />

• The incidence of diarrhea was higher in the cefdinir group.<br />

• Other adverse events were similar in both groups.<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefpodoxime = cefaclor<br />

• The bacteriologic eradication rates were similar in both<br />

119


Skin Structure Infection<br />

Tack, et Randomized,<br />

al. 16 double-mask,<br />

comparative,<br />

multicenter<br />

Tack, et<br />

al. 17<br />

Randomized,<br />

investigatorblind,<br />

multicenter<br />

• Patients aged 13<br />

years and older with a<br />

diagnosis of skin or<br />

structure infection<br />

including abscess,<br />

infected burn,<br />

carbuncle, cellulitis,<br />

infected dermatitis and<br />

trauma/surgical<br />

wounds.<br />

• Patients aged 6<br />

months to 12 years<br />

• Diagnosis of<br />

uncomplicated mild to<br />

moderate skin or skin<br />

structure infection<br />

warranting systemic<br />

antimicrobial therapy<br />

• Most common<br />

diagnoses were<br />

impetigo, infected<br />

dermatitis, wound<br />

infection and cellulitis<br />

952 • Cefdinir 300mg<br />

twice daily for 10 days<br />

• Cephalexin 500mg<br />

four times daily for 10<br />

days<br />

394 • Cefdinir 7mg/kg<br />

twice daily for 10 days<br />

• Cephalexin 10mg/kg<br />

four times daily for 10<br />

days<br />

posttherapy<br />

7-16 days<br />

posttherapy<br />

TOC =<br />

7-14 days<br />

LTFU =<br />

21-35 days<br />

posttherapy<br />

groups: 91% in cefpodoxime group and 92% in cefaclor group.<br />

• More bacterial isolates were susceptible in vitro to<br />

cefpodoxime (91%) than to cefaclor (84%).<br />

• The most common pretreatment isolates were H. influenzae,<br />

H. parainfluenzae and S. pneumoniae.<br />

Safety: cefpodoxime = cefaclor<br />

• Both treatments were well tolerated and had similar incidents<br />

of drug-related adverse events (cefpodoxime 11%, cefaclor 12%).<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefdinir = cephalexin<br />

• In the efficacy-assessable patients (~ 44%), clinical response<br />

rates were 88% in the cefdinir treatment group and 87% in<br />

cephalexin group.<br />

• The eradication rates were 93% in cefdinir group and 89% in<br />

cephalexin group.<br />

Safety: cefdinir < cephalexin<br />

• More patients reported adverse events in cefdinir group.<br />

(26% vs. 16%).<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefdinir = cephalexin<br />

• The clinical cure rates were 98.3% in the cefdinir group and<br />

93.8% in the cephalexin group. (P=0.056).<br />

• The microbiologic eradication rates were 99.4% in the<br />

cefdinir group and 97.4% in the cephalexin group.<br />

• The most common pathogens isolated were S. aureus and S.<br />

pyogenses.<br />

Safety: cefdinir = cephalexin<br />

• 16% of patients in the cefdinir group and 11% in the<br />

cephalexin group experienced adverse events.<br />

120


Stevens, et<br />

al. 18<br />

Randomized,<br />

double-blind,<br />

multicenter<br />

• Patients ≥ 12 years<br />

with acute, single site<br />

skin or skin-structure<br />

infections<br />

• Excluded patients<br />

with resistant<br />

pathogen.<br />

Uncomplicated Urinary Tract Infection (UTI)<br />

Leigh, et<br />

al. 19<br />

Randomized,<br />

double-blind<br />

parallel-group<br />

multicenter<br />

TOC= test of cure, LTFU=long term follow up.<br />

• Patents ≥ 13 years of<br />

age with symptoms of<br />

a urinary tract infection<br />

for more than 2 days<br />

• Patients who had a<br />

history of UTI, were<br />

hospitalized or were<br />

currently on other<br />

antibiotics were<br />

excluded<br />

371<br />

(271<br />

were<br />

evaluated)<br />

661<br />

(381<br />

were<br />

analyzed)<br />

• Cefpodoxime 400mg<br />

twice daily for 7-10<br />

days (matching cefaclor<br />

placebo)<br />

• Cefaclor 500mg<br />

three times daily for 7-<br />

10 days (with matching<br />

cefpodoxime placebo)<br />

• Cefdinir 100mg<br />

twice daily for 5 days<br />

• Cefaclor 250mg<br />

three times daily for 5<br />

days<br />

TOC =<br />

Day 7-10<br />

LTFU =<br />

2-3 weeks<br />

after<br />

treatment<br />

TOC = 5-9<br />

days<br />

LTFU = 6-<br />

8 weeks<br />

posttherapy<br />

• The most common adverse event was diarrhea.<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefpodoxime ≥ cefaclor<br />

• Both treatments were highly effective: 99% eradication rate<br />

and 86% cure rate.<br />

• Cefpodoxime had lower minimum inhibitor concentrations<br />

against the majority of Staphylococcus species than did cefaclor.<br />

• Cefaclor had a higher failure rate (4%) than cefpodoxime<br />

(1%).<br />

Safety: cefpodoxime = cefaclor<br />

• Both treatments were well tolerated.<br />

Primary Endpoints:<br />

• Clinical response rate<br />

• Bacteriologic eradication rate<br />

• Adverse events rate<br />

Efficacy: cefdinir = cefaclor<br />

• Clinical cure rates and microbiological eradication rates were<br />

equivalent (95% CI=-8.8% to 1.5% (cefdinir) and –1.8% to 15%<br />

(cefaclor)).<br />

• Resistance to cefaclor (6.7%) was significantly higher than<br />

cefdinir (3.7%). (P


Additional Evidence<br />

Dose Simplification: The oral cephalosporins are typically dosed twice daily, with a few<br />

exceptions (e.g. cephalexin). A study by Kardas et al. looked at patient adherence in respiratory<br />

tract infections with ceftibuten and other antibiotics. 20 The goal of the study was to establish<br />

whether dosing frequency (1 vs. 2 or 3 times daily) and other factors influenced compliance.<br />

Patients were randomized to ceftibuten 400mg daily or another antibiotic of the physician’s choice<br />

with a frequency of BID or TID. Compliance was measured in 406 patients by a pill count.<br />

Overall compliance was 76.6% with 97.6% for ceftibuten, 66.0% for antibiotics with BID dosing<br />

and 23.5% for antibiotics with TID dosing. Using a logistic regression analysis with a stepwise<br />

variable selection, dosing frequency was found to be a major variable associated with patient<br />

compliance (p = 0.00000, odds ratio 0.09, 95% confidence interval 0.057-0.165). This study,<br />

however did not measure an effect on outcome of the disease (respiratory tract infection<br />

improvement).<br />

A study by Tsi, et al looked at the efficacy and safety of once daily cefpodoxime proxetil<br />

suspension and cefaclor TID in the treatment of acute otitis media in children. 21 Satisfactory<br />

clinical outcome, either cure or improvement, was achieved at the end of treatment in 90% of<br />

patients in the cefaclor group and 95% of patients in the cefpodoxime group (p > 0.05). Clinical<br />

recurrence was identified at the follow-up visits in one case of the cefaclor group (3%), and none<br />

in the cefpodoxime group (p > 0.05). These drugs were well tolerated by 14/21 (67%) in the<br />

cefpodoxime-treated group and 27/32 (84%) in the cefaclor-treated group. The incidence of<br />

adverse events was slightly higher in the cefpodoxime group than in the cefaclor group, however<br />

the difference did not reach statistical significance (p > 0.05). In this study, cefaclor administered<br />

three times daily was as effective and safe as cefpodoxime administered once daily in the<br />

treatment of acute otitis media in children.<br />

Additionally, a study by de Klerk et al evaluated the efficacy of cefrtiaxone 1g QD to that of<br />

standard antibiotic treatment for lower respiratory tract infections. 22 Standard treatment was given<br />

according to guidelines from the American Thoracic Society. The mean duration of therapy was<br />

7.4 days in both groups. The average duration of hospitalization was 15.0 days for ceftriaxone and<br />

15.9 days for patients on standard therapy. Overall cure and improvement rates at the end of<br />

treatment were 47 (90%) for patients given ceftriaxone and 37 (77%) for patients receiving<br />

standard therapy. Once daily therapy with ceftriaxone was as effective as standard therapy in the<br />

treatment of lower respiratory infections, in view of multiple daily dosing regimens of most<br />

standard therapies.<br />

A study comparing IM ceftriaxone and oral amoxicillin-clavulanate in otitis media showed that the<br />

failure rate was similar in both groups (4.6% and 4.7%), respectively. 23 No significant differences<br />

in both groups were found in the dynamics of resolution of acute symptomatology, otoscopy<br />

findings, relapse rate at 30 days or tympanographic evidence of middle ear effusion at the<br />

scheduled visits. Recurrence rates of acute otitis media between days 31 and 90 were observed<br />

significantly in more children treated with amoxicillin/clavulanate than with cefriaxone (25 our of<br />

84 (29.4%) versus 11 out of 81 (13.6%) (P=0.012). A single ceftriaxone IM injection is as<br />

efficient as a 10-day oral amoxicillin clavulanate course and may be considered in the treatment of<br />

otitis media when children are unable to tolerate or absorb oral drugs, or in children refusing or<br />

unable to take oral therapy. Ceftriaxone is also be an option when compliance is questionable.<br />

Stable Therapy: Resistance to cephalosporins, especially the first generation cephalosporins, is a<br />

growing concern. Although changing between cephalosporins is not recommended, a different<br />

cephalosporin may be initiated due to therapeutic failure.<br />

Impact on Physician Visits: A literature search did not reveal clinical data pertinent to<br />

cephalosporin use and physician visits.


IX.<br />

Conclusions<br />

Not all cephalosporins have been directly compared to all other cephalosporins for certain<br />

indications. The clinical efficacy data presented above is a representative selection of recent<br />

comparative trials. These studies have shown comparable efficacy of some of the cephalosporins<br />

for the treatment of uncomplicated urinary tract infections, skin structure infections, and upper and<br />

lower respiratory tract infections. A few studies have shown greater efficacy of cephalosporin<br />

agents compared to amoxicillin/clavulanate and penicillin. Clinical data also demonstrates similar<br />

safety profiles between the cephalosporins (particularly within generations).<br />

In looking at indications, cephalexin is currently the only oral cephalosporin approved for bone<br />

infections caused by staphylococci or Proteus mirabilis. Likewise, cephalexin is also the only oral<br />

cephalosporin approved for genitourinary tract infections including acute prostatitis caused by E.<br />

coli, P. mirabilis, and Klebsiella Sp. Cefuroxime is the only cephalosporin approved for Lyme<br />

disease. Both of these agents are available with as generic formulations. Additionally, at least one<br />

agent in each cephalosporin generation is available in a generic formulation.<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the<br />

generics and OTC products in the class and offer no significant clinical advantage over other<br />

alternatives in general use.<br />

X. Recommendations<br />

No brand cephalosporin is recommended for preferred status.<br />

123


References<br />

1. Diagnosis and Management of Otitis Media. American Academy of Pediatrics, American<br />

Academy of Family Physicians. March 2004. Available at: www.aap.org. Accessed September 30,<br />

2004.<br />

2. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58,<br />

2004. Thomson PDR. Montvale, NJ. 2004.<br />

3. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

4. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

5. Clinical Pharmacology, 2004. Available on-line at: www.cp.gsm.com. Accessed September 21,<br />

2004.<br />

6. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

7. Briggs GG, Freeman RK, Yaffe SJ. Drugs in pregnancy and lactation. A reference guide to fetal<br />

and neonatal risk. Sixth Edition. Lippincott, Williams, & Wilkins, Philadelphia, 2002.<br />

8. Pessey J, Gehanno P, Thoroddsen E et al. Short course therapy with cefuroxime axetil for acute<br />

otitis media: results of a randomized multicenter comparison with amoxicillin/clavulanate. Pediatr<br />

Infect Dis J. 1999;18(10):854-859<br />

9. Nemeth MA, McCarthy J, Gooch WM et al. Comparison of cefdinir and penicillin for the<br />

treatment of streptococcal pharyngitis. Clin Ther. 1999;21(11):1873-1881.<br />

10. Pichichero ME, Gooch WM, Rodriguez W et al. Effective short-course treatment of acute group A<br />

beta-hemolytic streptococcal tonsillopharyngitis. Arch Pediatr Adolesc Med. 1994;148:1053-1060<br />

11. Asmar BI, Dajani AS, Del Beccaro MA et al. Comparison of cefpodoxime proxetil and cefixime in<br />

the treatment of acute otitis media in infants and children. Pediatrics. 1994;94:847-52<br />

12. Fogarty CM, Bettis RB, Griffin TJ et al. Comparison of a 5 day regimen of cefdinir with a 10 day<br />

regimen of cefprozil for treatment of acute exacerbations of chronic bronchitis. J Antimicrob<br />

Chemother. 2000;45:851-858<br />

13. Paster ZR, McAdoo AM, Keyserling CH et al. A comparison of five-day regimen of cefdinir with a<br />

seven-day regime of loracarbef of the treatment of acute exacerbations of chronic bronchitis. Int J Clin<br />

Pract. 2000; 54(5):293-299<br />

14. Drehobl M, Bianchi P, Constance H et al. Comparison of cefdinir and cefaclor in the treatment of<br />

community-acquired pneumonia. Antimicrobial Agents Chemother. 1997;41(7):1579-1583<br />

15. Phillips H, Van Hook CJ, Butler T et al. A comparison of cefpodoxime proxetil and cefaclor in the<br />

treatment of acute exacerbation of COPD in adults. Chest. 1993;104(5):1387-92<br />

16. Tack KJ, Littlejohn TW, Maillous G et al. Cefdinir versus cephalexin for the treatment of skin and<br />

skin-structure infections. Clin Ther. 1998;20(2):244-256.<br />

17. Tack KJ, Keyserling CH, McCarty J et al. Study of use of cefdinir versus cephalexin for treatment<br />

of skin infections in pediatric patients. Antimicrobial Agents Chemothe.. 1997;41(4):739-742<br />

18. Stevens DL, Pien F, Drehobl M. Comparison of oral cefpodoxime proxetil and cefaclor in the<br />

treatment of skin and soft tissue infections. Diagn Microbiol Infect Dis. 1993;16(2):123-129<br />

19. Leigh AP, Nemeth MA, Keyserling CH et al. Cefdinir versus cefaclor in the treatment of<br />

uncomplicated urinary tract infection. Clin Ther. 2000;22(7):818-825.<br />

20. Kardas P, Ratajczyk-Pakaluska E. Patient adherence in respiratory tract infections: ceftibuten<br />

versus other antibiotics. Pol Merkuriusz Lek 2001 Jun;10(60):445-9.<br />

21. Tsai HY, Huang LM, Chiu HH, et al. Comparison of once daily cefpodoxime proxetil suspension<br />

and thrice daily cefaclor suspension in the treatment of acute otitis media in children,. J Microbiol<br />

Immunol Infect 1998 Sep;31(3):165-70.<br />

22. de Klerk GJ, can Steijn JH, Lobatto S, et al. A randomized, multicenter study of ceftriaxone<br />

versus standard therapy in the treatment of lower respiratory tract infections. Int J Antimicrob Agents<br />

1999 Jul;12(2):121-7.<br />

23. Varsano I, Volovitz B, Horev Z, et al. Intramuscular ceftriaxone compared with oral<br />

amoxicillin/clavulanate for treatment of acute otitis media in children. Eur J Pediatr 1997<br />

Nov;156(11):858-63.<br />

124


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Miscellaneous β-Lactam Antibiotics<br />

Single Entity Agents<br />

AHFS 081207<br />

October 27, 2004<br />

I. Overview<br />

The β-lactam antibiotics can be classified into several groups according to their structural<br />

characteristics, but their unique structural feature is the presence of a four-membered β-lactam<br />

ring. The benefits of the β-lactam antibiotics have been known for 50 years and these antiinfectives<br />

continue to be beneficial due to their low toxicities and enhanced spectrum of activity.<br />

The nonpenicillin β-lactams exhibit a variable spectrum of antimicrobial activity and have a wide<br />

range of clinical uses. 1<br />

The carbapenems offer a broad antimicrobial spectrum, and meropenem has an improved safety<br />

profile compared with imipenem. Aztreonam is a useful alternative for patients with aerobic<br />

gram-negative infections who have allergies to the penicillins. 2<br />

Only a single agent, loracarbef, is available orally in this class, and only cefoxitin is available as a<br />

generic formulation. This review encompasses all dosage forms and strengths. Table 1 lists the<br />

drugs in this review.<br />

Table 1. Single Entity Miscellaneous β-Lactam Antibiotics in this <strong>Review</strong><br />

Generic Name β-Lactam Formulation Example Brand Name<br />

Classification<br />

Aztreonam Monobactam Injection Azactam<br />

Cefotetan Cephamycin Injection Cefotan<br />

Cefoxitin Cephamycin Injection Mefoxitin*<br />

Ertapenem Carbapenem Injection Invanz<br />

Loracarbef Carbacephem Oral Lorabid<br />

Meropenem Carbapenem Injection Merrem<br />

*Generic Available.<br />

II. Evidence Based Medicine and Current Treatment Guidelines<br />

The β-lactam antibiotics can be used for various infections. Because of this, an abundance of<br />

treatment guidelines are available that involve use of the β-lactam antibiotics. A review of some<br />

of the treatment guidelines and recommendations is highlighted in Table 2. Table 3 illustrates the<br />

spectrum of coverage for the drugs in this class.<br />

125


Table 2. Clinical Guidelines and Recommendations for the Miscellaneous β-Lactam Antibiotics<br />

Clinical Guideline<br />

Recommendation<br />

Hospital Pharmacist Consensus Reports: First-line treatment of patients with community-acquired<br />

The Antibiotic Selection for Community pneumonia (CAP) is oral azithromycin, with quinolone<br />

–Acquired Pneumonia Consensus Panel antibiotics as alternative first-line therapy.<br />

(The ASCAP Panel), 2002 3<br />

Severe CAP complicated by structural disease of the lung, and<br />

increased pseudomonas and polymicrobial infection, should<br />

be treated with cefepime plus levofloxacin plus/minus an<br />

aminoglycoside or ciprofloxacin plus an aminoglycoside plus<br />

azithromycin. Alternatively, CAP can be treated with<br />

ciprofloxacin plus cefepime plus azithromycin or a<br />

carbapenem plus azithromycin plus an aminoglycoside.<br />

Patients with severe pneumonia requiring ICU hospitalization<br />

may also be treated alternatively with a carbepenem plus an<br />

aminoglycoside plus azithromycin.<br />

Infectious Diseases Society of America: Initial empiric therapy involving drugs in this class for<br />

Guidelines for community-acquired suspected bacterial community-acquired pneumonia include:<br />

pneumonia in immunocompetent adults, • Inpatient, medical ward with no recent antibiotic<br />

2003 4<br />

therapy; give a quinolone alone or an advanced<br />

macrolide plus a β-lactam (cefotaxime, ceftriaxone,<br />

ampicillin-sulbactam, or ertapenem).<br />

• ICU, pseudomonas is not an issue; give a β-lactam<br />

plus either an advanced macrolide or a quinolone.<br />

• ICU, pseudomonas is an issue; give either (1) an<br />

antipseudomonal agent (piperacillin, piperacillintazobactam,<br />

imipenem, meropenem, or cefepime)<br />

plus ciprofloxacin or (2) an antipseudomonal agent<br />

plus an aminoglycoside plus a quinolone or a<br />

macrolide.<br />

• ICU, pseudomonas is an issue but patient has β-<br />

lactam allergy; give either (1) aztreonam plus<br />

levofloxacin or (2) aztreonam plus moxifloxacin or<br />

gatifloxacin, with or without an aminoglycoside.<br />

126


Table 3. Comparison of Bacterial Coverage of the Single Entity Miscellaneous β-Lactam Antibiotics 5<br />

Drug<br />

Spectrum<br />

Aztreonam The spectrum of aztreonam is limited to aerobic gram-negative bacteria; it has no<br />

gram-positive or anaerobic activity. Aztreonam may be used in place of an<br />

aminoglycoside since it is less nephrotoxic.<br />

Cefotetan<br />

Cefoxitin<br />

Ertapenem<br />

Gram-negative aerobes that are usually susceptible to aztreonam include: N.<br />

meningitidis, N. gonorrhoea, H. influenzae, Branhamella catarrhalis, Aeromonas<br />

hydrophila, Pasteurella multocida, and most Enterobactericeae (E.coli, Klebsiella,<br />

Citrobacter, Enterobacter, Morganella, Proteus, Providencia, Serratia, Salmonella,<br />

and Shigella). Aztreonam is often active against Pseudomonas aeruginosa, with<br />

potency slightly less than that of ceftazidime, although, as with all gram-negative<br />

organisms, specific susceptibility data should be considered. Anaerobes are not<br />

susceptible.<br />

Cefotetan differs from most other cephalosporins due to its activity against anaerobic<br />

organisms other than Bacteroides. It also has a somewhat longer half-life, allowing<br />

for twice-daily dosing. Cefotetan is active against most gram-positive bacteria other<br />

than enterococcus, although cefazolin, a first-generation agent, and cefmetazole,<br />

another second-generation cephamycin, are much more active against S. aureus than<br />

is cefotetan.<br />

Its gram-negative spectrum includes many Enterobacteriaceae, notably E. coli,<br />

Klebsiella, Proteus, and Providencia. Enterobacter, Serratia, and Pseudomonas are<br />

not susceptible. It also provides excellent coverage against N. gonorrhoeae and H.<br />

influenzae. Anaerobic coverage of cefotetan is similar to that of cefoxitin. Grampositive<br />

coverage includes nonpenicillinase- and penicillinase-producing<br />

Staphylococcus aureus (methicillin-resistant strains are resistant) and streptococci<br />

(except enterococci); however, its activity against these organisms is less than that of<br />

many other cephalosporins and penicillins.<br />

Cefoxitin has added stability against the β-lactamases, providing better resistance to<br />

certain gram-negative species. When it was marketed, cefoxitin was the first<br />

cephalosporin-like antibiotic to exhibit activity against anaerobes. Many institutions<br />

have substituted cefotetan for cefoxitin since their spectra of activity is extremely<br />

similar, and cefotetan can be dosed less frequently. In addition, cefoxitin has been<br />

found to be a potent inducer of β-lactamases.<br />

Cefoxitin possesses greater activity against gram-negative aerobes and anaerobes than<br />

currently available first-generation cephalosporins (at the time it became available).<br />

Due to its added stability against beta-lactamase-producing organisms, it is active<br />

against E. coli, Klebsiella, H. influenzae, Proteus, and Providencia. In addition, it<br />

provides excellent coverage against N. gonorrhoeae and is the drug of choice for the<br />

treatment of pelvic inflammatory disease (PID) due to N. gonorrhoeae. Cefoxitin has<br />

good activity against most gram-positive bacteria including nonpenicillinase- and<br />

penicillinase-producing Staphylococcus aureus (methicillin-resistant strains are not<br />

susceptible), streptococci (except enterococci), and many anaerobic bacteria<br />

(including B. fragilis).<br />

Ertapenem is highly stable against β-lactamases and has activity against a wide<br />

variety of gram-positive, gram-negative and anaerobic microorganisms, particularly<br />

the Enterobacteriaceae.<br />

The following organisms are generally considered susceptible to ertapenem in vitro<br />

and in clinical infections: Bacteroides distasonis; Bacteroides fragilis; Bacteroides<br />

ovatus; Bacteroides thetaiotaomicron; Bacteroides uniformis; Clostridium<br />

clostridioforme; Escherichia coli; Eubacterium lentum; Haemophilus influenzae<br />

(beta-lactamase negative); Klebsiella pneumoniae; Moraxella catarrhalis;<br />

Porphyromonas asaccharolytica; Prevotella bivia; Staphylococcus aureus (MSSA);<br />

Streptococcus agalactiae (group B streptococci); Streptococcus pneumoniae<br />

127


(penicillin susceptible strains); Streptococcus pyogenes (group A beta-hemolytic<br />

streptococci). Methicillin-resistant staphylococci and Enterococcus sp. are resistant<br />

to ertapenem. Limited in vitro activity has been demonstrated against Acinetobacter<br />

sp., Pseudomonas aeruginosa and Aeromonas sp. However, in vitro studies<br />

demonstrate that imipenem is three to ten times more potent against Acinetobacter sp.<br />

and Pseudomonas aeruginosa than ertapenem.<br />

Loracarbef<br />

Meropenem<br />

Note: On September 15, 2004, the FDA reported it had approved a supplemental new<br />

drug application for ertapenem for the addition of Staphylococcus epidermidis,<br />

Providencia rettgeri, Providencia stuartii, and Bacteroides vulgatus.<br />

The clinical utility and beta-lactamase stability of loracarbef is similar to that of the<br />

second-generation cephalosporins. Thus loracarbef's spectrum of activity includes<br />

pathogens frequently isolated in respiratory, urinary, skin, and soft-tissue infections.<br />

Gram-positive coverage includes Streptococcus pneumoniae; S. pyogenes; betahemolytic<br />

streptococci groups B, C, and G; Staphylococcus aureus (methicillinresistant<br />

strains are resistant); and Clostridium difficile. As with the cephalosporins,<br />

loracarbef is inactive against enterococci. Gram-negative coverage includes activity<br />

against E. coli, Neisseria gonorrhoeae, N. meningitidis, Haemophilus influenzae,<br />

Moraxella catarrhalis, Klebsiella pneumoniae, Helicobacter pylori, and P. mirabilis.<br />

In vitro susceptibility testing has shown variable results with different inoculum sizes<br />

of beta-lactamase-positive strains of H. influenzae, M. catarrhalis, Staphylococcus<br />

aureus, and E. coli. Inoculum size does not affect loracarbef's activity against<br />

β-lactamase-negative strains.<br />

The spectrum of activity of meropenem is very similar to imipenem although<br />

meropenem is more active against Enterobacteriaciae, Haemophilus influenzae,<br />

gonococcus, and Pseudomonas aeruginosa.<br />

128


III.<br />

Comparative Indications of the Single Entity Miscellaneous β-Lactam Antibiotics


Aztreonam<br />

✔<br />

Complicated<br />

and noncomplicated,<br />

pyelonephritis<br />

and cystitis<br />

✔<br />

Pneumonia<br />

and<br />

bronchitis<br />

Skin and<br />

Skin<br />

Structure<br />

Infections<br />

Table 4. FDA-Approved Indications for the Single Entity Misc. B-Lactams 5-8<br />

Drug UTI Lower<br />

RTI*<br />

Upper<br />

RTI*<br />

Septicemia<br />

Intra-<br />

Abdominal<br />

Infections<br />

Gynecologic<br />

Infections<br />

Surgery<br />

Concurrent<br />

Initial<br />

Therapy<br />

✔ ✔ ✔ ✔ ✔ ✔<br />

With grampositive<br />

coverage<br />

Bone and<br />

Joint<br />

Infections<br />

Perioperative<br />

Prophylaxis<br />

Cefotetan ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔<br />

Caused by<br />

S. aureus<br />

Cefoxitin ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔<br />

Pneumonia<br />

and lung<br />

abscesses<br />

Ertapenem ✔<br />

Complicated,<br />

including<br />

pyelonephritis<br />

✔<br />

Complicated<br />

✔<br />

Complicated<br />

✔<br />

Acute pelvic<br />

infections:<br />

postpartum<br />

endomyometritis<br />

and septic<br />

abortion<br />

Loracarbef<br />

✔<br />

Uncomplicated<br />

cystitis and<br />

pyelonephritis<br />

✔<br />

Pneumonia<br />

and<br />

bronchitis<br />

✔<br />

Otitis media,<br />

sinusitis,<br />

pharyngitis,<br />

tonsillitis<br />

✔<br />

Uncomplicated<br />

Meropenem ✔<br />

Complicated<br />

appendicitis<br />

and<br />

peritonitis<br />

*Respiratory Tract Infection<br />

Community-<br />

Acquired<br />

Pneumonia<br />

✔<br />

With<br />

concurrent<br />

bacteremia<br />

Bacterial<br />

Meningitis<br />

✔<br />

Pediatric<br />

patients ≥ 3<br />

months<br />

only


IV.<br />

Pharmacokinetic Parameters<br />

Table 5. Pharmacokinetic Parameters of the Single Entity Miscellaneous β-Lactams 5-8<br />

Drug Mechanism of Action Bioavailability Protein<br />

Binding<br />

Metabolism<br />

Aztreonam Aztreonam is mainly Poorly absorbed from 56-60% 6-16% in the<br />

bactericidal and inhibits the GI tract<br />

liver<br />

the third and final stage<br />

of bacterial cell wall<br />

synthesis by<br />

preferentially binding to<br />

specific penicillinbinding<br />

proteins (PBPs)<br />

that are located inside the<br />

bacterial cell wall.<br />

Cefotetan<br />

Cefoxitin<br />

Ertapenem<br />

Cefotetan inhibits the<br />

third and final stage of<br />

bacterial cell wall<br />

synthesis by<br />

preferentially binding to<br />

specific penicillinbinding<br />

proteins (PBPs)<br />

that are located inside the<br />

bacterial cell wall. The<br />

drug’s ability to interfere<br />

with PBP-mediated cell<br />

wall synthesis ultimately<br />

leads to cell lysis. Lysis<br />

is mediated by bacterial<br />

cell wall autolytic<br />

enzymes.<br />

Cefoxitin, a beta-lactam<br />

cephamycin similar to<br />

penicillins and<br />

cephalosporins, inhibits<br />

the third and final stage<br />

of bacterial cell wall<br />

synthesis by<br />

preferentially binding to<br />

specific penicillinbinding<br />

proteins (PBPs)<br />

that are located inside the<br />

bacterial cell wall.<br />

Ertapenem exhibits<br />

bactericidal activity due<br />

inhibition of cell wall<br />

synthesis mediated via<br />

binding to penicillin<br />

binding proteins (PBPs).<br />

Ertapenem inhibits the<br />

third and final stage of<br />

bacterial cell wall<br />

synthesis by<br />

preferentially binding to<br />

specific penicillinbinding<br />

proteins (PBPs)<br />

that are located inside the<br />

bacterial cell wall.<br />

Poorly absorbed from<br />

the GI tract; peak levels<br />

occur in 1.5-3 hours<br />

following IM injection<br />

Peak levels occur in<br />

20-30 minutes following<br />

an IM dose<br />

75-90% No 1-10% of a<br />

dose is<br />

present in<br />

plasma and<br />

urine as an<br />

active<br />

tautomer of<br />

the drug<br />

50-80% 2%<br />

metabolized<br />

90% 95% Yes,<br />

hydrolysis<br />

Active Elimination Half-Life<br />

Metabolites<br />

No 60-70% Renal 1.7 hours<br />

No<br />

No<br />

Renal,<br />

49-81% of<br />

dose is<br />

excreted in<br />

urine<br />

Excreted<br />

unchanged in<br />

by the<br />

kidneys<br />

80% renal,<br />

10% feces<br />

3-4.5<br />

hours<br />

40-60<br />

minutes<br />

4.5 hours


Loracarbef<br />

Meropenem<br />

By preferentially binding<br />

to specific penicillinbinding<br />

proteins (PBPs)<br />

located inside the<br />

bacterial cell wall,<br />

loracarbef inhibits the<br />

third and final stage of<br />

bacterial cell wall<br />

synthesis.<br />

Meropenem inhibits cell<br />

wall formation,<br />

facilitates bacterial cell<br />

lysis, and is mainly<br />

bactericidal. It inhibits<br />

the third and final stage<br />

of bacterial cell wall<br />

synthesis by<br />

preferentially binding to<br />

specific penicillinbinding<br />

proteins (PBPs)<br />

that are located inside the<br />

bacterial cell wall.<br />

90% absorbed following<br />

oral administration;<br />

administration of tablets<br />

with food decreases the<br />

peak plasma<br />

concentrations by<br />

40-50% and delays time<br />

to peak concentrations<br />

by 30-60 minutes; the<br />

extent of absorption is<br />

about the same for both<br />

formulations, but the<br />

increased rate of<br />

absorption and resulting<br />

increases in peak serum<br />

concentrations from the<br />

suspension should be<br />

considered when<br />

substituting capsules for<br />

the suspension. The<br />

capsules should not be<br />

used in place of the<br />

suspension for otitis<br />

media.<br />

Mean peak plasma<br />

concentrations at the end<br />

of a 30-minute IV<br />

infusion of a single dose<br />

in normal volunteers are<br />

approximately<br />

23 mcg/ml (range<br />

14-26 mcg/ml) for the<br />

500 mg dose and<br />

49 mcg/ml (range<br />

39-58 mcg/ml) for the 1<br />

gram dose.<br />

25% No evidence<br />

of metabolism<br />

2% Minimally<br />

metabolized<br />

No Renal 1 hour<br />

No<br />

70% is<br />

excreted<br />

unchanged in<br />

the urine<br />

1.2 hours<br />

V. Drug Interactions<br />

Table 6 lists the most significant drug-drug interactions for the drugs indexed by Drug Interactions<br />

Facts (cefotetan and cefoxitin). 9 Interactions for the other drugs in the class are described below.<br />

Table 6. Drug Interactions of the Single Entity Miscellaneous β-Lactams 9<br />

Drug Significance Interaction Mechanism<br />

Cephalosporins<br />

(cefotetan)<br />

Level 2 Cefotetan and ethanol (or drugs<br />

that contain ethanol)<br />

Aldehyde dehydrogenase inhibition by methyltetrazolethiol<br />

results in acetaldehyde accumulation. Occurrence and severity<br />

are unpredictable. Dose-response and length of enzyme<br />

inhibition are unknown. A disulfiram-like reaction manifested<br />

by flushing, tachycardia, bronchospasm, sweating, nausea and<br />

vomiting may occur when ethanol is ingested after a patient has<br />

been on cefotetan, or other cephalosporins with the<br />

methyltetrazolethiol moiety (cefmandole, cefoperazone,<br />

Cephalosporins<br />

(cefotetan and<br />

cefoxitin)<br />

Cephalosporins<br />

(cefotetan and<br />

cefoxitin)<br />

Level 2<br />

Level 2<br />

Cefotetan, cefoxitin and<br />

aminoglycosides<br />

Cefotetan, cefoxitin and<br />

anticoagulants (anisindione,<br />

warfarin, dicumarol)<br />

cefonicid, ceforanide, and moxalactam).<br />

Mechanism is unknown. Nephrotoxicity may be<br />

increased. Bactericidal activity against certain pathogens<br />

may be enhanced.<br />

Mechanism is unknown. The anticoagulant effect of warfarin<br />

is increased.<br />

132


Aztreonam 8<br />

Precipitant<br />

drug<br />

Probenecid<br />

Furosemide<br />

Antibiotics<br />

(e.g., cefoxitin,<br />

imipenem)<br />

*<br />

Additional Drug-Drug Interactions for the Single Entity Miscellaneous β-Lactam Antibiotics<br />

Object drug *<br />

Aztreonam<br />

Aztreonam<br />

Description<br />

Concomitant administration causes clinically insignificant increases in<br />

aztreonam serum levels.<br />

Antibiotics may induce high levels of -lactamase in vitro in some gramnegative<br />

aerobes such as Enterobacter and Pseudomonas sp, resulting in<br />

antagonism to many -lactam antibiotics including aztreonam. Do not use -<br />

lactamase-inducing antibiotics concurrently with aztreonam.<br />

Aztreonam Aminoglycosides If an aminoglycoside is used concurrently with aztreonam, especially if high<br />

dosages of the former are used or if therapy is prolonged, monitor renal function<br />

because of potential nephrotoxicity and ototoxicity of aminoglycoside<br />

antibiotics.<br />

= Object drug increased. = Object drug decreased.<br />

Ertapenem<br />

When ertapenem is coadministered with probenecid (500mg by mouth every 6 hours), probenecid competes<br />

6, 7, 8<br />

for active tubular secretion and reduces the renal clearance of ertapenem. Based on total ertapenem<br />

concentrations, probenecid increased the AUC by 25% and reduced the plasma and renal clearances by<br />

20% and 35%, respectively. The half-life increased from 4 to 4.8 hours. Because of the small effect on halflife,<br />

the coadministration with probenecid to extend the half-life of ertapenem is not recommended.<br />

In vitro studies indicate ertapenem does not inhibit P-glycoprotein-mediated transport of digoxin or<br />

vinblastine and ertapenem is not a substrate for P-glycoprotein-mediated transport. In vitro studies in human<br />

liver microsomes indicate that ertapenem does not inhibit metabolism mediated by any of the following six<br />

cytochrome P450 isoforms: 1A2, 2C9, 2C19, 2D6, 2E1 and 3A4. Drug interactions caused by inhibition of<br />

P-glycoprotein-mediated drug clearance or CYP-mediated drug clearance with the listed isoforms are<br />

unlikely.<br />

Other than with probenecid, no specific clinical drug interaction studies have been conducted.<br />

Loracarbef<br />

Probenecid: As with other β-lactam antibiotics, renal excretion of loracarbef is inhibited by probenecid and<br />

resulted in an approximate 80% increase in the AUC for loracarbef. 11<br />

Meropenem<br />

Probenecid competes with meropenem for active tubular secretion and thus inhibits the renal excretion of<br />

meropenem. 6-8 This led to statistically significant increases in the elimination half-life (38%) and in the extent<br />

of systemic exposure (56%). Therefore, the coadministration of probenecid with meropenem is not<br />

recommended.<br />

There is evidence that meropenem may reduce serum levels of valproic acid to subtherapeutic levels<br />

(therapeutic range considered to be 50 to 100micrograms/ml total valproate).<br />

133


VI. Adverse Drug Events of the Single Entity Miscellaneous β-Lactam<br />

Antibiotics<br />

Table 7. Common Adverse Events (%) Reported for the Single Entity Misc. B-Lactam Antibiotics 6, 8<br />

Adverse Event Aztreonam Cefotetan Cefoxitin Ertapenem Loracarbef Meropenem<br />

Body as a Whole<br />

Malaise


VII.<br />

Dosing and Administration for the Single Entity Miscellaneous β-Lactam<br />

Antibiotics<br />

Table 8. Dosing for the Single Entity Miscellaneous β-Lactam Antibiotics<br />

Drug Availability Dose /Frequency/Duration<br />

Aztreonam Powder for injection:<br />

500mg, 1g, and 2g<br />

Give IM or IV. Individualize dosage.<br />

Aztreonam Dosage Guide (Adults)<br />

Type of infection Dose 1 Frequency (hours)<br />

Urinary tract infection 500mg or 1g 8 or 12<br />

Moderately severe systemic infections 1 or 2g 8 or 12<br />

Severe systemic or life-threatening<br />

infections<br />

2g 6 or 8<br />

1 Maximum recommended dose is 8 g/day.<br />

Aztreonam Dosage Guide (Children)<br />

Type of infection Dose 1 Frequency (hours)<br />

Mild-to-moderate infections 30mg/kg 8<br />

Moderate-to-severe infections 30mg/kg 6 or 8<br />

1 Maximum recommended dose is 120mg/kg/day.<br />

Duration:<br />

Therapy duration depends on the severity of infection. Generally, continue<br />

aztreonam for ≥48 hours after the patient becomes asymptomatic or evidence<br />

of bacterial eradication has been obtained. Persistent infections may require<br />

treatment for several weeks. Do not use doses smaller than those indicated.<br />

Cefotetan<br />

Powder for injection:<br />

1, 2, and 10g<br />

Injection: 1g/50ml<br />

and 2g/50ml<br />

Adults:<br />

The usual dosage is 1 or 2g IV or IM every 12 hours for 5 to 10 days.<br />

General Cefotetan Dosage Guidelines<br />

Type of Infection Daily Dose Frequency and Route<br />

Urinary tract<br />

1 to 4g 500mg every 12hrs IV or IM<br />

1 or 2g every 24hrs IV or IM<br />

1 or 2g every 12hrs IV or IM<br />

Skin/Skin structure<br />

Mild-to-moderate 2<br />

2g<br />

2g q 24hrs IV<br />

1g q 12hrs IV or IM<br />

Severe 4g 2g q 12 hrs IV<br />

Other Sites 2 to 4g 1 or 2g every 12hrs IV or IM<br />

Severe 4g 2g every 12hrs IV<br />

Life-threatening 6g 1 3g every 12hrs IV<br />

1 Maximum daily dosage should not exceed 6g.<br />

2 Klebsiella pneumoniae skin and skin structure infections should be treated with 1 or 2 g every<br />

12 hours IV or IM.<br />

Prophylaxis:<br />

To prevent postoperative infection in clean contaminated or potentially<br />

contaminated surgery in adults, give a single 1 or 2g IV dose 0.5 to 1 hour<br />

prior to surgery. In patients undergoing cesarean section, give the dose as<br />

soon as the umbilical cord is clamped.<br />

135


Cefoxitin<br />

Powder for injection:<br />

1, 2, and 10g<br />

Injection: 1g/50ml<br />

and 2g/50ml<br />

Adults:<br />

The dosage range is 1 to 2g every 6 to 8 hours. Maintain antibiotic therapy<br />

for group A β-hemolytic streptococcal infections for ≥10 days to guard<br />

against the risk of rheumatic fever or glomerulonephritis.<br />

Cefoxitin Dosage Guidelines<br />

Type of infection Daily dosage Frequency and route<br />

Uncomplicated (pneumonia, 3 to 4g 1g every 6 to 8 hours IV<br />

urinary tract, cutaneous) 1<br />

Moderately severe or severe 6 to 8g 1g every 4 hours or 2g<br />

every 6 to 8 hours IV<br />

Infections commonly requiring<br />

higher dosage (e.g. gas<br />

gangrene)<br />

12g<br />

1 Including patients in whom bacteremia is absent or unlikely.<br />

2g every 4 hours or 3g<br />

every 6 hours IV<br />

Ertapenem<br />

Loracarbef<br />

Powder for injection:<br />

1g<br />

Oral pulvules:<br />

200mg and 400mg<br />

Oral suspension:<br />

100mg/5ml (100ml),<br />

200mg/5ml (100ml)<br />

strawberry bubble<br />

gum flavor<br />

Uncomplicated gonorrhea: 2g IM with 1g oral probenecid given concurrently<br />

or ≤30 minutes before cefoxitin.<br />

Prophylactic use, surgery: Administer 2g IV 30 to 60 minutes prior to surgery<br />

followed by 2g every 6 hours after the first dose for ≤24 hours.<br />

Prophylactic use, cesarean section: Administer 2g IV as soon as the umbilical<br />

cord is clamped. If a three-dose regimen is used, give the second and third 2g<br />

dose IV, 4 and 8 hours after the first dose.<br />

Prophylactic use, transurethral prostatectomy: administer 1g prior to surgery;<br />

1g every 8 hours for ≤5 days.<br />

Dose: 1g given once a day.<br />

Ertapenem may be administered by IV infusion for up to 14 days or IM<br />

injection for up to 7 days. When administered IV, infuse ertapenem over a<br />

period of 30 minutes.<br />

IM administration of ertapenem may be used as an alternative to IV<br />

administration in the treatment of those infections for which IM therapy is<br />

appropriate.<br />

Dosage/Duration of Loracarbef<br />

Population/Infection<br />

Dosage<br />

(mg)<br />

Duration<br />

(days)<br />

Adults ≥13 years of age<br />

Lower respiratory tract<br />

Secondary bacterial infection of acute 200-400 q 12 hrs 7<br />

bronchitis<br />

Acute bacterial exacerbation of<br />

400 q 12 hrs 7<br />

chronic bronchitis<br />

Pneumonia 400 q 12 hrs 14<br />

Upper respiratory tract<br />

Pharyngitis/Tonsillitis 200 q 12 hrs 10 1<br />

Sinusitis 400 q 12 hrs 10<br />

Skin and skin structure<br />

136


Meropenem<br />

Powder for injection:<br />

500mg, 1g, also Add-<br />

Vantage vials in<br />

500mg and 1g<br />

Uncomplicated 200 q 12 hrs 7<br />

Urinary tract<br />

Uncomplicated cystitis 200 q 24 hrs 7<br />

Uncomplicated pyelonephritis 400 q 12 hrs 14<br />

Infants and children(6 months to 12<br />

years) 2<br />

Upper respiratory tract<br />

Acute otitis media 3<br />

Acute maxillary sinusitis<br />

Pharyngitis/Tonsillitis<br />

Skin and skin structure<br />

Impetigo<br />

30mg/kg/day in<br />

divided doses q 12 hrs<br />

15mg/kg/day in<br />

divided doses q 12 hrs<br />

15mg/kg/day in<br />

divided doses q 12 hrs<br />

1 In treatment of infections caused by S. pyogenes, administer for ≥10 days.<br />

2 Do not exceed adult recommended doses.<br />

3 Use suspension; it is more rapidly absorbed than capsules, resulting in higher peak plasma<br />

concentrations when given at the same dose.<br />

Weight<br />

10<br />

10 1<br />

Loracarbef Pediatric Suspension Dosage<br />

Daily dose 15mg/kg/day Daily dose 30mg/kg/day<br />

100mg/5 ml<br />

twice daily<br />

200mg/5ml<br />

twice daily<br />

100mg/5 ml<br />

twice daily<br />

7<br />

200mg/5ml<br />

twice daily<br />

lb kg ml tsp ml tsp ml tsp ml Tsp<br />

15 7 2.6 0.5 -- -- 5.2 1 2.6 0.5<br />

29 13 4.9 1 2.5 0.5 9.8 2 4.9 1<br />

44 20 7.5 1.5 3.8 0.75 -- -- 7.5 1.5<br />

57 26 9.8 2 4.9 1 -- -- 9.8 2<br />

Adults:<br />

1g by IV administration every 8 hours. Give over 15 to 30 minutes or as an<br />

IV bolus injection (5 to 20ml) over 3 to 5 minutes.<br />

Use in pediatric patients:<br />

For pediatric patients from ≥3 months of age, the dose is 20 or 40mg/kg<br />

every 8 hours (maximum dose is 2g every 8 hours), depending on the type of<br />

infection (intra-abdominal or meningitis). Administer pediatric patients<br />

weighing > 50kg 1g every 8 hours for intra-abdominal infections and 2g<br />

every 8 hours for meningitis. Give over 15 to 30 minutes or as an IV bolus<br />

injection (5 to 20 ml) over 3 to 5 minutes.<br />

Recommended Meropenem IV Dosage Schedule for Pediatrics with<br />

Normal Renal Function<br />

Type of infection Dose (mg/kg) Dosing interval<br />

Intra-abdominal 20 every 8 hours<br />

Meningitis 40 every 8 hours<br />

137


Special Dosing Considerations<br />

Table 9. Special Dosing Considerations for the Single Entity Miscellaneous β-Lactams 5-9<br />

Drug Renal Dosing Hepatic Pediatric Use Pregnancy Can Drug Be Crushed/Stability<br />

Dosing<br />

Category<br />

Aztreonam<br />

Yes<br />

Patients with Cr. Cl<br />

10-30ml/min<br />

reduce dose by<br />

50%, after 1 or 2g<br />

loading dose.<br />

No Children only; there is<br />

insufficient data<br />

regarding IM<br />

administration to<br />

pediatric patients or<br />

dosing in pediatric<br />

patients with renal<br />

impairment.<br />

B Use solutions for IV infusion at<br />

concentrations ≤2% w/v within<br />

48 hours following constitution if<br />

kept at controlled room temperature<br />

(15° to 30°C; 59° to 86°F) or within<br />

7 days if refrigerated (2°to 8°C; 36°<br />

to 46°F).<br />

Cefotetan<br />

Cefoxitin<br />

Ertapenem<br />

Yes<br />

Increase the<br />

frequency of<br />

administration.<br />

Yes<br />

Patients with Cr. Cl<br />

≤50ml/min should<br />

have the frequency<br />

of administration<br />

increased.<br />

Yes<br />

Patients with a Cr.<br />

Cl ≤30ml/min and<br />

end-stage renal<br />

disease should<br />

receive a dose of<br />

500mg daily.<br />

No<br />

No<br />

No<br />

Safety and efficacy in<br />

children have not been<br />

established.<br />

Safety and efficacy of<br />

pediatric patients from<br />

birth to three months<br />

of age has not been<br />

established. In<br />

patients three months<br />

or older, higher doses<br />

of the drug have been<br />

associated with an<br />

increased incidence of<br />

eosinophilia and<br />

elevated SGOT.<br />

Safety and efficacy in<br />

pediatric patients has<br />

not been established;<br />

use in patients under<br />

18 years of age is not<br />

recommended.<br />

B Cefotetan maintains potency for 24<br />

hours at room temperature (25°C;<br />

77°F), for 96 hours refrigerated<br />

(5°C; 40°F) and for 1 week frozen.<br />

After reconstitution and subsequent<br />

storage in disposable glass or plastic<br />

syringes, cefotetan is stable for 24<br />

hours at room temperature and 96<br />

hours refrigerated.<br />

B<br />

B<br />

Reconstituted vials maintain<br />

potency for six hours at room<br />

temperature or an additional 48<br />

hours under refrigeration. Further<br />

dilution maintains potency for an<br />

additional 18 hours at room<br />

temperature or an additional 48<br />

hours if refrigerated.<br />

Do not mix or co-infuse ertapenem<br />

with other medications. Do not use<br />

diluents containing dextrose.<br />

The reconstituted solution may be<br />

stored at room temperature and used<br />

within 6 hours or stored for 24<br />

hours under refrigeration and used<br />

within 4 hours after removal from<br />

refrigeration. Solutions should not<br />

be frozen.<br />

Loracarbef<br />

Yes<br />

Patients with Cr. Cl<br />

between 10-<br />

49ml/min = ½ the<br />

recommended dose,<br />

Cr. Cl


Meropenem<br />

Yes<br />

Reduce dosage in<br />

patients with a Cr.<br />

Cl


VIII. Comparative Effectiveness of the Single Entity Miscellaneous β-Lactams<br />

Clinical trials confirm that multiple cephalosporins (cefprozil, cefpodoxime, cefixime, loracarbef,<br />

and ceftibuten) are as effective as traditional therapies for the management of acute otitis media,<br />

pharyngitis/tonsillitis, sinusitis, bronchitis, pneumonia, urinary tract infections, and skin and skinstructure<br />

infections. 12 Selection of a specific agent is influenced by susceptibility data and safety,<br />

as well as issues of compliance. Table 10 describes published, peer-reviewed trials of the drugs<br />

within this class.<br />

Table 10. Additional Outcomes Evidence for the Single Entity Miscellaneous β-Lactams<br />

Study Sample Treatment /<br />

Results<br />

Description<br />

General Comparative Class Studies<br />

In vitro study in<br />

multidrugresistant<br />

Ps.<br />

Seven<br />

strains of the<br />

bacteria<br />

aeruginosa 13<br />

All strains of Ps.<br />

Aeruginosa were<br />

resistant to piperacillin,<br />

The goal of this study was to examine the combined effects of antibiotic<br />

combinations by agar incorporation inhibitory tests and by time-kill tests on seven<br />

isolates of multidrug-resistant Ps. aeruginosa. Results indicated:<br />

meropenem,<br />

ceftazidime,<br />

cefoperazonesulbactam,<br />

• Among the two-drug combinations, the combination aztreonam and<br />

amikacin was the most effective, inhibiting proliferation in five of the<br />

seven strains.<br />

aztreonam,<br />

amikacin, and<br />

ciprofloxacin<br />

• Among the three-drug combinations, the combinations of piperacillin,<br />

ceftazidime and amikacin, and that of ceftazidime, aztreonam and amikacin<br />

were the most effective, inhibiting proliferation in all seven strains.<br />

• In the killing tests, the three-drug combination of ceftazidime, aztreonam<br />

and amikacin was the most effective. This three-drug combination had<br />

bacteriostatic effects on all seven strains 2, 4, 6 and 24 hours after drug<br />

addition, synergic effects on 2-3 strains and bactericidal effects on<br />

1-2 strains after 4, 6 and 24 hours.<br />

• Summary: The three-drug combination of ceftazidime, aztreonam and<br />

amikacin may be effective against P. aeruginosa resistant to all commonly<br />

used antipseudomonal drugs, and deserves further study.<br />

A surveillance<br />

study of 65 labs<br />

in the United<br />

States 14 - From 1998-2001 A surveillance study of 65 laboratories in the United States, looking at Pseudomonas<br />

aeruginosa and Acinetobacter baumannii showed:<br />

• Greater than 90% of isolates of Ps. aeruginosa from hospitalized patients<br />

were found to be susceptible to amikacin and piperacillin-tazobactam; 80<br />

to 90% of isolates to be susceptible to cefepime, ceftazidime, imipenem,<br />

and meropenem; and 70 to 80% of isolates to be susceptible to<br />

ciprofloxacin, gentamicin, levofloxacin, and ticarcillin-clavulanate.<br />

• From 1998 to 2001, decreases in antimicrobial susceptibility (percents)<br />

among non-intensive-care-unit (non-ICU) inpatients and ICU patients,<br />

respectively, were greatest for ciprofloxacin (6.1 and 6.5), levofloxacin<br />

(6.6 and 3.5), and ceftazidime (4.8 and 3.3).<br />

• Combined 1998 to 2001 results for A. baumannii isolated from non-ICU<br />

inpatients and ICU patients, respectively, demonstrated that >90% of<br />

isolates tested were susceptible to imipenem (96.5 and 96.6%) and<br />

meropenem (91.6 and 91.7%); fewer isolates from both non-ICU inpatients<br />

and ICU patients were susceptible to amikacin and ticarcillin-clavulanate<br />

(70 to 80% susceptible); and


Report from the 3,047<br />

MYSTIC<br />

bacterial<br />

Program 15 isolates<br />

collected in<br />

2002 from<br />

16 North<br />

America<br />

sites<br />

The Meropenem Yearly<br />

Susceptibility Test<br />

Information Collection<br />

Program<br />

The activity of meropenem and nine broad-spectrum antibiotics were assessed<br />

against 3,047 bacterial isolates:<br />

• The overall rank order of susceptibility of the 10 antimicrobial agents<br />

tested against Gram-negative isolates was: meropenem (98%) > imipenem<br />

(97%) > cefepime (95%) > tobramycin (93%) > piperacillin/tazobactam =<br />

gentamicin (92%) > ceftazidime (91%) > ciprofloxacin (87%) > aztreonam<br />

(86%) > ceftriaxone (74%).<br />

• The utility of meropenem against Pseudomonas aeruginosa isolates has<br />

increased steadily with a rise in percent susceptibility each year from<br />

78.2% in 1999 to a present rate of 93.1% susceptible.<br />

• The susceptibility for ciprofloxacin against the same P. aeruginosa isolates<br />

decreased from 82.9 to 72.3% susceptible over four years.<br />

• Carbapenem resistance remains rarely documented and these betalactamase-stable<br />

agents appear to be an alternative treatment option for<br />

serious community-acquired or nosocomial infections in high risk patient<br />

populations.<br />

Report from the<br />

SENTRY<br />

antimicrobial<br />

surveillance<br />

Program 16<br />

Report from the<br />

SENTRY<br />

antimicrobial<br />

surveillance<br />

Program 17<br />

Sustained<br />

activity and<br />

spectrum<br />

against<br />

Enterobacter<br />

spp. and<br />

Klebsiella<br />

spp.<br />

Geographic<br />

variations in<br />

activity<br />

against Ps.<br />

aeruginosa<br />

Meta-analysis of n=79<br />

randomized trials randomized<br />

of antibiotic clinical trials<br />

therapy in intraabdominal<br />

infections 18<br />

- In evaluating the rates and trend resistance to extended-spectrum beta-lactams and<br />

other antimicrobial agents to Enterobacter spp. and Klebsiella spp. isolated between<br />

1997 and 2000:<br />

• Among Enterobacter spp., resistance (MIC>or=32 mg/l) to aztreonam,<br />

ceftazidime and ceftriaxone ranged from 12.3 to 21.2% over the 4 years,<br />

whereas resistance in Klebsiella (MIC>or=2 mg/l) ranged from 5.9 to<br />

6.8%.<br />

• Carbapenems (imipenem, meropenem) and cefepime had excellent activity<br />

against both ceftazidime-susceptible and -resistant Enterobacter spp. and<br />

Klebsiella spp. (>99% susceptible), although the minimum inhibitory<br />

concentration values of cefepime were higher in ceftazidime-resistant<br />

isolates compared with ceftazidime-susceptible isolates.<br />

- In comparing the activities of cefepime and eight other β-lactams against 6,969<br />

isolates of Ps. aeruginosa collected during 1997-2000:<br />

• The beta-lactams exhibited a wide range of potency, with carbapenems<br />

most active (meropenem, 80-91% susceptible; imipenem, 76-88%<br />

susceptible).<br />

• Piperacillin/tazobactam was the most active penicillin (77-80%<br />

susceptible), and cefepime (67-83% susceptible) had an average 2% (range,<br />

0.7-3.5%) greater susceptibility rate than ceftazidime (66-80% susceptible)<br />

across all regions.<br />

• The rank order of beta-lactam activity according to percent resistant<br />

isolates in North American Ps. aeruginosa strains was: meropenem (4.8%<br />

resistant) > cefepime (6.8%) > imipenem (8.6%) > piperacillin/tazobactam<br />

(10.3%) > piperacillin (12.9%). Only 2.3% and 6.5% of isolates were<br />

resistant to amikacin or tobramycin, respectively, and nearly 16% of Ps.<br />

aeruginosa strains were resistant to ciprofloxacin.<br />

• Compared with other geographic regions, strains of P. aeruginosa remain<br />

most susceptible in North America.<br />

- In reviewing the published randomized studies looking at antibiotic use in intraabdominal<br />

infections:<br />

• Studies are limited due to a lack of disease severity stratification and a<br />

relatively small study population for most antibiotics.<br />

• The clinical success rate of the antibiotics studied in a larger population is<br />

comparable for gentamicin + clindamycin (80%), tobramycin +<br />

clindamycin (83%), meropenem (89%), imipenem (85%), aztreonam +<br />

clindamycin (89%), cefoxitin (88%), cefotetan (92%), moxalactam* (83%),<br />

cefotaxime + metronidazole (87%), ampicillin/sulbactam (87%).<br />

Piperacillin/tazobactam has in most studies a success rate of approximately<br />

90%.<br />

• The aggregated data on adverse events and clinical failure rate do not show<br />

a major advantage for any of these antibiotics.<br />

• The clinical success rate of the best-studied antibiotics is similar and the<br />

choice which antibiotic is used depends on the expected pathogens and the<br />

resistance rate in a clinical setting.<br />

141


Aztreonam Clinical Evidence<br />

In vitro synergy<br />

of aztreonam and<br />

quinolones<br />

40 strains of<br />

Ps.<br />

aeruginosa<br />

Broth microdilution was<br />

used to determine MICs<br />

Because aztreonam can be used in combination with other antibiotics to enhance<br />

antimicrobial spectrum and produce synergistic activity, the drug was studied in vitro<br />

with ciprofloxacin, gatifloxacin, levofloxacin, cefepime, ceftazidime, and imipenem:<br />

against gramnegative<br />

bacilli 19<br />

and Enterobacteriaceae<br />

• No antagonism or indeterminate interactions were identified between any<br />

of the combinations.<br />

• The overall rate of synergy or partial synergy for aztreonam with<br />

fluoroquinolone combinations was 63.4% versus P. aeruginosa, greatest<br />

for aztreonam with gatifloxacin (67.5%).<br />

• Aztreonam with ceftazidime or cefepime versus P. aeruginosa had<br />

75.0 - 85.0% partial or complete synergy rates, but aztreonam with<br />

imipenem showed dominant indifference (65.0%).<br />

• In contrast, aztreonam with imipenem was more likely to exhibit synergy<br />

(32.5%) when tested against Enterobacteriaceae.<br />

• Summary: Aztreonam, often used as an aminoglycoside substitute in<br />

antimicrobial combinations, continues to demonstrate enhanced, but<br />

variable drug activity interactions for contemporary antimicrobial<br />

combinations when tested against recent (2002) clinical isolates.<br />

The role of<br />

- reviewing the literature to determine the clinical efficacy of aztreonam:<br />

surgery patients 20 with the aminoglycosides.<br />

aztreonam in<br />

nosocomial<br />

pneumonia in<br />

• Numerous studies have documented that aztreonam’s effectiveness is equal<br />

of superior to that of other suitable antibiotics in the treatment of<br />

nosocomial pneumonia.<br />

critically ill<br />

• Aztreonam’s safety profile makes the drug especially attractive compared<br />

• Due to bacterial resistance with use of broader-spectrum alternatives,<br />

aztreonam is a sound choice as well for the treatment of nosocomial<br />

Cefepime vs.<br />

ticarcillin and<br />

clavulanate<br />

potassium plus<br />

aztreonam for<br />

febrile<br />

neutropenia 21 n=126 Open label,<br />

randomized,<br />

comparative trial<br />

Aztreonam vs.<br />

ampicillin,<br />

cotrimoxazole,<br />

minocycline,<br />

nalidixic acid,<br />

norfloxacin,<br />

ciprofloxacin,<br />

gentamicin, or<br />

ceftriaxone in the<br />

treatment of<br />

urinary tract<br />

infections 22<br />

Aztreonam plus<br />

clindamycin vs.<br />

tobramycin plus<br />

clindamycin for<br />

abdominal<br />

n=342 gramnegative<br />

isolates<br />

An in vitro study of the<br />

listed drugs’ efficacy on<br />

E. coli, Klebsiella<br />

pneumoniae, proteus<br />

species, and Ps.<br />

aeruginosa<br />

infections 23 n=70 The average length of<br />

treatment was 10 days<br />

(tobramycin and<br />

clindamycin group) and<br />

9 days (aztreonam and<br />

clindamycin group)<br />

pneumonia.<br />

In order to evaluate the efficacy of empiric applied cefepime (C) as monotherapy<br />

versus combination therapy consisting of ticarcillin and clavulanate potassium and<br />

aztreonam (T/A), patients were randomized to treatment:<br />

• Using afebrile status following 3 days of therapy as a primary endpoint,<br />

both regimens produced comparable clinical response rates (C = 55% vs.<br />

T/A = 61%).<br />

• Also, the use of vancomycin for resistant gram-positive infections and<br />

alteration of gram-negative infection coverage was similar in both groups<br />

(C = 40% vs. T/A = 47% and C = 29% vs. T/A = 24%).<br />

• Both treatment groups had similar needs for empirical antifungal therapy<br />

(C = 25% vs. T/A = 22%).<br />

• Treatment of febrile neutropenic patients with these two treatment<br />

regimens offers similar efficacy results.<br />

In evaluating the efficacy of aztreonam versus eight other antibiotic treatments for<br />

bacteria common to urinary tract infections:<br />

• Out of 342 isolates, 76.6% were E. coli, 14.3% were Klebsiella<br />

pneumoniae, 5.2 were proteus spp., and 3.8 were Ps. aeruginosa.<br />

• The aztreonam showed 78% sensitivity against gram-negative bacilli,<br />

which is better than norfloxacin, which showed 62.2% sensitivity.<br />

Patients with intraabdominal infections were randomly assigned in a double-blinded<br />

manner to receive tobramycin plus clindamycin (TM/C) or aztreonam plus<br />

clindamycin (AZ/C). Results indicated:<br />

• In approximately one-half of both groups, the source of infection was<br />

perforated colon or perforated appendix.<br />

• Clinical response to therapy did not differ, as 84% of the TM/C patients<br />

and 78% of the AZ/C patients had satisfactory clinical responses.<br />

• The two regimens differed in adverse effects, as an elevated PT/PTT was<br />

more frequently (p < 0.05) observed in AZ/C. All PT/PTT elevations<br />

responded to injections of vitamin K, and no serious bleeding occurred.<br />

• Choice between these regimens depends on differences in adverse effects,<br />

142


Randomized,<br />

comparative<br />

study of<br />

cefoxitin,<br />

cefotetan, and<br />

cefalotin* (1 st<br />

generation<br />

cephalosporin not<br />

available in the<br />

United States) 24 n=155 Randomized treatments<br />

included: Cefoxitin 3 x<br />

1g (14 subjects),<br />

Cefotetan 2 x 1g (43<br />

subjects), Cefalotin 3 x<br />

2g for 24 hours (27<br />

subjects), Cefalotin 3 x<br />

2g for 72 hours (41<br />

subjects) and a control<br />

group of 30 subjects<br />

without prophylaxis<br />

Retrospective<br />

analysis of<br />

antibiotic<br />

prophylaxis<br />

(cefotetan vs.<br />

cefoxitin)<br />

following<br />

cesarean section 25 n=385 Single doses of<br />

cefotetan 1g vs.<br />

cefoxitin 2g<br />

Randomized,<br />

double-blind,<br />

controlled study<br />

of ertapenem vs.<br />

piperacillintazobactam<br />

for<br />

skin/skin<br />

structure<br />

infections 26 n=146 Ertapenem 1g QD vs.<br />

piperacillin-tazobactam<br />

13.5g divided Q6H,<br />

duration infection<br />

dependent<br />

Randomized,<br />

double-blind<br />

study of<br />

ertapenem vs.<br />

piperacillintazobactam<br />

for<br />

methicillinsusceptible<br />

Staphylococcal<br />

aureas in<br />

skin/skin<br />

structure<br />

infections 27 n=529 Ertapenem 1g QD vs.<br />

piperacillin-tazobactam<br />

3.375g Q6H, duration<br />

infection dependent<br />

as both regimens appear equally effective for treatment of abdominal<br />

infections in conjunction with appropriate surgical intervention.<br />

Cefotetan and Cefoxitin Clinical Evidence<br />

In evaluating cefotetan, cefoxitin, and cefalotin efficacy in perioperative prophylaxis<br />

in abdominal gynecological prophylaxis:<br />

• No patients receiving any prophylaxis developed infectious complications,<br />

while such complications occurred in 10% of the control without<br />

prophylaxis.<br />

• Antibiotic prophylaxis effectively prevented the infectious complications<br />

and shortened the postoperative hospital stay.<br />

• Despite its narrower anti-bacterial spectrum cefalotin is not inferior in its<br />

efficacy to the second and the third-generation cephalosporins.<br />

• The longer 72-hour cefalotin scheme shows no advantages to the 24-hour<br />

frequency.<br />

In comparing single-dose antibiotic prophylaxis in various subpopulations based on<br />

risk factors for postsurgical infection following cesarean section:<br />

• Postsurgical infection rate was greater in non-elective cases, 7.4%, vs.<br />

elective cases, 3.0% (p =0.056) as was the rate of endometritis (3.2% vs.<br />

1.2%, p =0.185).<br />

• No differences were noted based on antibiotic regimen.<br />

• Postsurgical infection rate was greater for 28 cases at high risk for both<br />

surgical infection and endometritis (17.9%) when compared to all 357<br />

other cases (4.5%), p =0.003. No difference was noted for endometritis.<br />

• Of the 28 cases 28.6% of patients treated with cefoxitin and 7.1% of cases<br />

treated with cefotetan developed postsurgical infection (p =0.13).<br />

• Summary: Overall cefoxitin and cefotetan provided equivalent clinical<br />

outcome. A small subset of patients with multiple risk factors for infection<br />

may benefit from cefotetan.<br />

Ertapenem Clinical Evidence<br />

During a clinical trial of complicated skin/skin structure infections in patients who<br />

received outpatient antimicrobial therapy (OPAT) with either ertapenem or<br />

piperacillin-tazobactam:<br />

• In evaluable patients managed by OPAT, 45 (83.3%) of 54 treated with<br />

ertapenem and 41 (82.0%) of 50 treated with piperacillin-tazobactam were<br />

cured at the test of cure assessment 10-21 days post-therapy (OR 1.2 (95%<br />

CI, 0.4-3.2), p=0.78).<br />

• The safety profile of both drugs was generally similar; diarrhea was the<br />

most common adverse event in both groups.<br />

• Summary: Among patients who received OPAT, ertapenem 1g daily was<br />

as effective as piperacillin-tazobactam 3.375g Q6H.<br />

In this subset study, the efficacy of ertapenem was compared with piperacillintazobactam<br />

for the treatment of complicated skin/skin structure infections caused by<br />

methicillin-susceptible S. aureus (MSSA):<br />

• At the test of cure assessment 10-21 days post-therapy, 54 of 67 (80.6%)<br />

protocol evaluable patients in the ertapenem group and 55 of 68 (80.9%) in<br />

the piperacillin-tazobactam group were cured (odds ratio: 1.0 (95%<br />

confidence interval (CI): 0.4-2.4), p = 0.99).<br />

• In both treatment groups, cure rates were higher in patients with<br />

monomicrobial than polymicrobial infections, but the difference was not<br />

significant.<br />

• Summary: Therapy with ertapenem 1g daily was as effective as<br />

piperacillin-tazobactam 13.5g divided in four daily doses, in this subgroup<br />

analysis of patients with MSSA.<br />

Two randomized,<br />

double-blind,<br />

multicenter trial<br />

of ertapenem vs.<br />

ceftriaxone in<br />

UTI 28 n=480 Ertapenem 1g QD vs.<br />

ceftriaxone 1g QD;<br />

patients could switch to<br />

oral therapy after ≥3<br />

days of IV study<br />

therapy<br />

In treating complicated urinary tract infections in adults with ertapenem or<br />

ceftriaxone:<br />

• Ninety-six percent of these patients were switched to oral therapy, usually<br />

ciprofloxacin; the median (range) duration of parenteral and total therapy,<br />

respectively, was 4 (2-14) days and 13 (14-18) days for ertapenem and 4<br />

(2-14) days and 13 (3-17) days for ceftriaxone.<br />

• At the primary efficacy endpoint 5-9 days after treatment, 229 (89.5%)<br />

patients who received ertapenem and 204 (91.1%) patients who received<br />

143


ceftriaxone had a favorable microbiological response (95% confidence<br />

interval, -7.4 to 4.0), indicating that outcomes in the two treatment groups<br />

were equivalent.<br />

• Success rates in both treatment groups were similar when compared by<br />

stratum and severity of infection.<br />

• The frequency and severity of drug-related adverse events were generally<br />

similar in both treatment groups.<br />

Post-hoc analysis<br />

of data from three<br />

large randomized,<br />

double-blind<br />

trials of<br />

ertapenem vs.<br />

piperacillintazobactam<br />

n=1,558 Ertapenem 1g QD vs.<br />

piperacillin-tazobactam<br />

3.375g Q6H; duration<br />

infection dependent<br />

In vitro<br />

n=482<br />

susceptibility of strains of<br />

antibioticresistant<br />

urinary pathogens<br />

urinary<br />

pathogens to<br />

ertapenem and 12<br />

other antiibotics 30<br />

Susceptibility testing of<br />

identified pathogens to<br />

ertapenem, ampicillin,<br />

cefazolin, cefuroxime,<br />

cefotaxime,<br />

amoxicillin/clavulanate<br />

acid,<br />

piperacillin/tazobactam,<br />

imipenem, gentamicin,<br />

amikacin, fosfomycin,<br />

ciprofloxacin, and cotrimoxazole<br />

Prospective,<br />

randomized,<br />

double-blind<br />

multicenter<br />

comparison of<br />

ertapenem vs.<br />

ceftriaxone in<br />

CAP 31 n=364 Ertapenem 1g QD vs.<br />

ceftriaxone 1g QD;<br />

durations of parenteral<br />

and total therapy were<br />

5.5 and 11.5 days for<br />

ertapenem and 5.6 and<br />

11.7 for ceftriaxone<br />

In evaluating the efficacy of ertapenem versus piperacillin-tazobactam for the<br />

treatment of polymicrobial complicated intra-abdominal, complicated skin/skin<br />

structure infections, and acute pelvic infections:<br />

• At the test-of-cure assessment, there were no significant differences in<br />

outcomes between the two treatment groups for any of the three infections.<br />

• Cure rates (clinical and microbiological for intra-abdominal infection,<br />

clinical for skin/skin-structure and pelvic infections) in microbiologically<br />

evaluable patients for ertapenem and piperacillin-tazobactam, respectively,<br />

were 85.6% (154/180 evaluable patients) and 82.5% (127/154) for<br />

polymicrobial intra-abdominal infection, 80.3% (53/66) and 78.7% (48/61)<br />

for polymicrobial skin/skin-structure infection, and 95.7% (88/92) and<br />

92.6% (88/95) for polymicrobial pelvic infection.<br />

• Respective cure rates for all evaluable patients in the original trials were:<br />

83.6% and 80.4% for intra-abdominal, 83.9% and 85.3% for skin/skinstructure,<br />

and 93.9% and 91.5% for pelvic infections.<br />

• Summary: These data show that in the three trials, ertapenem 1g once a<br />

day was highly effective for the treatment of polymicrobial infections and<br />

as effective as piperacillin-tazobactam 3.375g every 6 hours.<br />

In testing the in vitro susceptibility of urinary pathogens to that of the drugs listed in<br />

the previous column:<br />

• The distribution of pathogens tested was as follows: Escherichia coli (n =<br />

315), Proteus mirabilis (n = 42), Klebsiella spp. (n = 14) and AmpCproducing<br />

Enterobacteriaceae (n = 111).<br />

• All the strains were susceptible to ertapenem, imipenem and amikacin.<br />

• The MIC(90) of ertapenem ranged from a minimum of 0.03mg/L for<br />

Proteus vulgaris and a maximum of 1mg/L for Enterobacter spp.<br />

Ertapenem was the most active of all drugs tested in all cases.<br />

• On comparing antibiotic resistance among ESBL-producing strains of E.<br />

coli (n = 35) and E. coli strains not producing ESBLs (n = 280),<br />

statistically significant differences were obtained for ciprofloxacin (P =<br />

0.002) and gentamicin (p = 0.011).<br />

• Regarding ertapenem, only a slight increase in MIC(50) was seen, the<br />

value being 0.015mg/L for strains not producing ESBLs versus 0.03mg/L<br />

for ESBL-producing strains.<br />

In order to compare the efficacy and safety of ertapenem and ceftriaxone for the<br />

treatment of hospitalized adult patients with serious community-acquired pneumonia<br />

(CAP) requiring IV therapy:<br />

• Streptococcus pneumoniae was the most frequently isolated pathogen in<br />

both treatment groups.<br />

• Cure rates were 92.2% for clinically evaluable patients in the ertapenem<br />

group and 93.6% for those in the ceftriaxone group (95% CI for the<br />

difference, adjusted for stratum, -8.6 to 5.7), fulfilling the criteria for<br />

statistical equivalence.<br />

• At completion of parenteral therapy, 94.7% of patients in the ertapenem<br />

group and 95.8% in the ceftriaxone group showed clinical improvement.<br />

• Infused vein complications (ertapenem, 3.4% [8/236]; ceftriaxone, 7.3%<br />

[9/123]) and elevated transaminase levels (ertapenem, 6.3% [13/207];<br />

ceftriaxone, 7.1% [8/113]) were the most common adverse events in both<br />

groups.<br />

• Summary: In this study, ertapenem therapy, with an oral switch option,<br />

was as effective as ceftriaxone with the same oral switch option for<br />

treatment of CAP requiring initial parenteral therapy. The overall safety<br />

profiles of the two drugs were comparable.<br />

144


Randomized,<br />

double-blind,<br />

multicenter study<br />

comparing<br />

loracarbef vs.<br />

doxycycline for<br />

acute bacterial<br />

maxillary<br />

sinusitis 32 n=662 Loracarbef 400mg BID<br />

or doxycycline 200mg x<br />

1 followed by 100mg<br />

QD, for 10 days<br />

Loracarbef vs.<br />

cefaclor and<br />

norfloxacin in the<br />

treatment of<br />

uncomplicated<br />

pyelonephritis 33 n=245 Loracarbef 400mg BID<br />

vs. cefaclor 500mg<br />

TID, and loracarbef<br />

400mg BID vs.<br />

norfloxacin 400mg<br />

BID; treatment was<br />

continued for ≥14 days<br />

Randomized,<br />

double-blind,<br />

parallel study of<br />

loracarbef vs.<br />

penicillin V 34 n=344 Ten day therapy with<br />

loracarbef (200mg BID<br />

or 15mg/kg/day oral<br />

suspension in two<br />

divided doses up to a<br />

maximum of<br />

375mg/day) vs.<br />

penicillin V (250mg<br />

QID or 20mg/kg/day<br />

suspension in four<br />

divided doses up to a<br />

maximum of<br />

500mg/day)<br />

Randomized, n=214<br />

double-blind trial children<br />

of loracarbef vs. ages 6<br />

cefaclor in months to 12<br />

pediatric patients years<br />

with skin and<br />

skin structure<br />

infections 35<br />

Oral suspensions given<br />

for 7 days at<br />

15mg/kg/day in two<br />

divided doses for<br />

loracarbef and<br />

20mg/kg/day in three<br />

divided doses for<br />

cefaclor<br />

Loracarbef Clinical Evidence<br />

In evaluating the efficacy of loracarbef vs. doxycycline in the treatment of acute<br />

maxillary sinusitis:<br />

• Streptococcus pneumoniae and/or Haemophilus influenzae were isolated<br />

from approximately 75% of patients.<br />

• The clinical response rate (cure or improvement) was significantly higher<br />

for patients receiving loracarbef (98.2%) than for those who received<br />

doxycycline (92.2%).<br />

• There was no significant difference between the two groups with respect to<br />

bacteriological outcome, although more of the pre-treatment isolates were<br />

resistant to doxycycline (35 strains) than to loracarbef (five strains).<br />

• Adverse events related to the gastrointestinal tract occurred in 11.7% and<br />

10.6% of loracarbef- and doxycycline-treated patients respectively; therapy<br />

was terminated prematurely in ten patients in the loracarbef group and in<br />

nine in the doxycycline group.<br />

Because conventional oral antibiotics such as the sulfonamides and the<br />

aminopenicillin are limited by development of resistance, the therapeutic efficacy of<br />

loracarbef was compared with cefaclor and norfloxacin in two trials:<br />

• Escherichia coli was the causative pathogen in 85.0% of these patients.<br />

• Successful post therapy clinical and bacteriologic responses were similar<br />

for all three study drugs: 94.1 and 86.8%, 96.0 and 80.0%, 97.7 and 88.4%<br />

for loracarbef, cefaclor, and norfloxacin, respectively.<br />

• Late post therapy clinical responses were 87.4, 83.3, and 91.7% for the<br />

loracarbef, cefaclor, and norfloxacin groups, respectively. Bacteriologic<br />

responses for the three groups were 79.6, 60.0, and 88.9%.<br />

• The most frequent adverse effects (headache, diarrhea, and nausea) were<br />

experienced by three patients (2.5%) in the loracarbef group; headaches<br />

were noted in two (4.7%) cefaclor patients, and diarrhea was noted in three<br />

(7.0%) patients in the cefaclor group. Gastrointestinal events were noted in<br />

four patients (4.8%) in the norfloxacin group.<br />

• Summary: Loracarbef is comparable in efficacy and safety to both cefaclor<br />

and norfloxacin as oral therapy for uncomplicated pyelonephritis.<br />

In the treatment of Group A beta-hemolytic streptococcal (GABHS) pharyngitis and<br />

tonsillitis, therapeutic efficacy of loracarbef and penicillin V was evaluated:<br />

• Post-therapy clinical responses were similar for evaluable patients in both<br />

treatment groups: 97.4% of the loracarbef group (101/115 patients cured<br />

and 11/115 improved) and 96.0% of the penicillin group (101/124 patients<br />

cured and 18/124 improved).<br />

• A statistically significant difference in the pathogen elimination rate was<br />

noted between treatment groups: post-therapy throat cultures were negative<br />

for GABHS in 94.8% (109/115) of loracarbef-treated patients compared<br />

with 87.1% (108/124) of penicillin-treated patients (p = 0.040).<br />

• Loracarbef and penicillin V were comparable in terms of safety.<br />

• Summary: Loracarbef twice daily is more effective in eradicating GABHS<br />

than penicillin V four times daily, and the two drugs are comparable in<br />

safety.<br />

In comparing the safety and efficacy of loracarbef and cefaclor for the treatment of<br />

skin and skin structure infections in children:<br />

• Assessment 72 hours after completion of the 7-day course of treatment<br />

indicated a favorable clinical response plus eradication of the pretherapy<br />

pathogen in 97.3% of the 74 loracarbef-treated patients eligible for<br />

evaluation and 92.3% of 78 evaluable cefaclor-treated patients.<br />

• Favorable response rates at a second post treatment visit 10 to 14 days after<br />

the end of therapy were 95.6% in 68 evaluable loracarbef-treated patients<br />

and 86.2% in 65 treated with cefaclor.<br />

• The incidence of adverse reactions, including gastrointestinal effects, was<br />

low in both groups.<br />

• No statistical difference in clinical or bacteriologic efficacy or safety was<br />

detected between patients treated with loracarbef and cefaclor.<br />

145


In vitro activity<br />

and<br />

pharmacodynami<br />

cs of oral betalactam<br />

antibiotics 36 - MICs were determined<br />

for penicillin,<br />

amoxicillin,<br />

amoxicillin-clavulanic<br />

acid, cefprozil,<br />

cefuroxime,<br />

cefpodoxime, cefaclor,<br />

and loracarbef<br />

Two multicenter,<br />

randomized,<br />

single-masked,<br />

trial of loracarbef<br />

vs. clarithromycin<br />

in children with<br />

acute otitis<br />

media 37 n=334 Loracarbef 15mg/kg or<br />

clarithromycin<br />

7.5mg/kg orally BID for<br />

10 days<br />

Randomized,<br />

double-blind,<br />

multicenter trial<br />

of cefdinir vs.<br />

loracarbef for<br />

exacerbations of<br />

chronic<br />

bronchitis 38 n=586 Cefdinir 300mg BID for<br />

five days or loracarbef<br />

400mg BID for seven<br />

days<br />

In evaluating the frequency and reduced susceptibility to penicillin, and to compare<br />

the in vitro activity and pharmacodynamics of oral β-lactam antibiotics against<br />

clinical isolates of Streptococcus pneumoniae:<br />

• The frequency of penicillin nonsusceptible S. pneumoniae was 28.7%.<br />

• For 25 penicillin-intermediate isolates, amoxicillin and amoxicillinclavulanic<br />

acid were significantly more active than cefprozil, cefaclor, and<br />

loracarbef.<br />

• The T > MIC (average time above MIC) for amoxicillin and amoxicillinclavulanic<br />

acid, simulated at 13.3mg/kg every 8 hours, was significantly<br />

longer than that for all other beta-lactams.<br />

• Summary: Amoxicillin and amoxicillin-clavulanic acid have superior in<br />

vitro activity and longer T > MIC for penicillin-intermediate isolates than<br />

the other oral beta-lactams.<br />

In order to evaluate loracarbef with clarithromycin with regard to efficacy,<br />

tolerability, and patient acceptance, two studies were performed in children aged 6<br />

months to 3 years:<br />

• The combined results of these two studies showed that the efficacy and<br />

tolerability of loracarbef were comparable to those of clarithromycin.<br />

• Adverse events were reported by 46.4% of loracarbef patients and 41.0%<br />

of clarithromycin patients, with no statistically significant difference<br />

between groups.<br />

• In the intent-to-treat analysis, 57.9% of loracarbef patients were cured at<br />

the termination of the study, compared with 55.7% of clarithromycin<br />

patients.<br />

• Improvement was seen in 4.1% of loracarbef patients and 2.7% of<br />

clarithromycin patients.<br />

• Results of the patient acceptance survey showed a clear preference for<br />

loracarbef over clarithromycin.<br />

• Difficulties with administration of treatment were reported by 36.3% of<br />

clarithromycin caregivers, compared with 7.8% of loracarbef caregivers<br />

(p < 0.001).<br />

• A desire to stop treatment was reported by 23.8% of clarithromycin<br />

caregivers, compared with 7.8% of loracarbef caregivers (p < 0.001).<br />

• Taste and texture issues were most frequently cited as reasons for<br />

nonacceptance.<br />

In comparing the clinical efficacy of cefdinir versus loracarbef for the treatment of<br />

chronic bronchitis:<br />

• The clinical cure rates were 86% (138/160) and 85% (141/166) for the<br />

evaluable patients treated with cefdinir and loracarbef, respectively.<br />

• Respiratory tract pathogens were isolated from 457 (78%) of 586<br />

admission sputum specimens, with the predominant pathogens being<br />

Haemophilus parainfluenzae, H. influenzae, Moraxella catarrhalis and<br />

Staphylococcus aureus.<br />

• The microbiological eradication rates at the test-of-cure visit were 88%<br />

(193/219 pathogens) and 90% (227/251 pathogens) for the evaluable<br />

patients treated with cefdinir and loracarbef, respectively.<br />

• Adverse event rates while on treatment were 30% and 21% for cefdinirand<br />

loracarbef-treated patients, respectively.<br />

Blinded taste<br />

comparison of<br />

twelve common<br />

pediatric drugs 39 - - In evaluating the taste of twelve antimicrobial suspensions with regards to smell,<br />

texture, taste, aftertaste, and overall acceptance:<br />

• Overall, loracarbef scored highest but not significantly higher than Keflex,<br />

Suprax and Ceclor, all of which score higher than the other test drugs.<br />

• Cefzil and Augmentin scored just below this group of drugs and higher<br />

than all other test drugs.<br />

• Vantin was inferior to these drugs primarily because of its low score in<br />

aftertaste. It was ranked along with V-Cillin-K, Veetids, Sulfatrim and<br />

Pediazole, the lowest scoring group of drugs other than Dynapen, which<br />

scored lower than all other test drugs.<br />

• No difference overall was detected between the two penicillin VK<br />

suspensions evaluated, V-Cillin-K and Veetids.<br />

146


n=900<br />

Meropenem Clinical Evidence<br />

In vitro activity<br />

- To compare the in vitro activity of meropenem with that of other agents with a broadspectrum<br />

of meropenem<br />

and other broadspectrum<br />

antibiotics 40 isolates<br />

(including<br />

multiresistant,<br />

of antibacterial activity:<br />

• The potency and spectrum<br />

and anaerobes, were undoubtedly<br />

beta-lactamases.<br />

of the carbapenems, unequalled against aerobes<br />

influenced by their stability to serine<br />

grampositive<br />

aerobes,<br />

• Meropenem and imipenem exhibited essentially the same spectrum of<br />

activity but imipenem was often less potent, notably against Gram-negative<br />

aerobes, including Pseudomonas aeruginosa and Burkholderia cepacia.<br />

Enterobacteriaceae,<br />

and, to some extent, the newer cephalosporins (MIC90s 0.06--8mg/L) and<br />

• Conversely, the activity of the third-generation (MIC90s 0.016--64 mg/L)<br />

nonfermenters,<br />

many genera of Enterobacteriaceae because of chromosomally mediated<br />

the augmented penicillins (MIC90s 1 to >128mg/L) was unreliable against<br />

and<br />

enzymes or the, now commonplace, plasmid-mediated beta-lactamases.<br />

anaerobes<br />

• Ciprofloxacin had modest activity (MIC90s 1--64mg/L) against Grampositive<br />

aerobes, was potent against nutritionally fastidious species, had<br />

again variable activity against the Enterobacteriaceae (MIC90s 0.008--<br />

4mg/L) and was inactive against many strains of Pseudomonas,<br />

Burkholderia and Acinetobacter, resulting in MIC90s of 4 to >128mg/L.<br />

• The aminoglycosides were impressive only against the Enterobacteriaceae,<br />

with amikacin alone active (MIC90s 2--8mg/L) against the 11 species<br />

tested.<br />

• Summary: The study suggests also that, judged on activity in vitro,<br />

meropenem or imipenem are the only monotherapy options for empirical<br />

antibacterial therapy of polymicrobic infections or when local<br />

epidemiology indicates the predominance of multiresistant<br />

Enterobacteriaceae. Instability to current and emerging beta-lactamases is<br />

progressively compromising the use of all other beta-lactam compounds.<br />

Stability of<br />

antipseudomonal<br />

β-lactams with<br />

portable<br />

elastomeric<br />

pumps 41 - Aztreonam (100 g/liter),<br />

piperacillin (128 g/liter,<br />

with tazobactam),<br />

azlocillin (128 g/liter),<br />

mezlocillin (128<br />

g/liter), ceftazidime<br />

The stability of antipseudomonal beta-lactams in concentrated solutions was<br />

examined in view of their potential administration by continuous infusion with<br />

external pumps in patients with cystic fibrosis:<br />

• Aztreonam, piperacillin /tazobactam, and azlocillin* remained 90% stable<br />

for up to more than 24 hours at 37 degrees C (mezlocillin was stable at<br />

degrees C but not at 37 degrees C).<br />

25<br />

(120 g/liter),<br />

cefpirome* (32 g/liter),<br />

and cefepime (50<br />

g/liter)<br />

• Ceftazidime, cefpirome*, and cefepime (50 g/liter) remained 90% stable<br />

for up to 24, 23.7, and 20.5 hours at 25 degrees C but only for 8, 7.25, and<br />

13 hours at 37 degrees C, respectively. The control of temperature<br />

therefore appears to be critical for all three cephalosporins that cannot be<br />

recommended for use in portable pumps carried under clothes for<br />

prolonged periods for reasons of stability.<br />

• Imipenem and meropenem were too unstable (10% degradation at 25<br />

degrees C after 3.5 and 5.15h, respectively) to be recommended for use by<br />

Meropenem vs.<br />

ceftazidime<br />

monotherapy for<br />

cancer patients<br />

with chemoinduced<br />

febrile<br />

neutropenia 42 n=99 Duration dependent on<br />

response and severity of<br />

neutropenia<br />

Randomized,<br />

controlled trial of<br />

meropenem<br />

versus imipenem/<br />

cilastatin 43 n=182 Meropenem 500mg<br />

Q12 (or 1g Q12 if<br />

necessary) or<br />

imipenem/cilastatin<br />

500mg Q12 (or 1g Q12<br />

if necessary), for a<br />

duration of 7-14 days<br />

continuous infusion.<br />

Patients (hospitalized for fever > 38 degrees C and ANC ≤ 500/cmm) were<br />

prospectively treated with meropenem, while their outcome was compared to patients<br />

treated with ceftazidime for the same indication:<br />

• Duration of fever, duration of neutropenia, and number of patients with<br />

pyrexia of undetermined origin were also similar.<br />

• Therapeutic outcome was the same between the two groups.<br />

• Eighty-four percent of patients receiving meropenem and 79% receiving<br />

ceftazidime required no modification of the initially assigned therapeutic<br />

regimen.<br />

In evaluating the efficacy and safety of meropenem versus imipenem/cilastatin in<br />

patients with lower respiratory tract infections, urinary tract infections, and other<br />

infections:<br />

• The overall efficacy rates were 90% for the meropenem group and 87% for<br />

the imipenem/cilastatin group, and the bacterial eradication rates were 86%<br />

in both groups.<br />

• The adverse drug reaction rates were 9.7% and 8.6%, respectively.<br />

• The results showed that there were no statistical differences between these<br />

two groups (p > 0.05).<br />

147


Randomized,<br />

controlled trial of<br />

meropenem vs.<br />

imipenem in the<br />

prophylactic<br />

treatment of<br />

septic<br />

complications of<br />

acute<br />

pancreatitis 44 n=176 Meropenem 500mg TID<br />

vs. imipenem 500mg<br />

QID<br />

In vitro activity - Multidrug-resistant Ps.<br />

ciprofloxacin 45<br />

of combo therapy<br />

aeruginosa isolates<br />

with cefepime,<br />

were studied by the<br />

piperacillin/tazob<br />

time-kill method<br />

actam, or<br />

meropenem with<br />

In looking at the efficacy of meropenem and imipenem in the treatment of severe<br />

acute pancreatitis:<br />

• No difference was observed between patients treated with meropenem and<br />

those treated with imipenem in terms of incidence of pancreatic infection<br />

(11.4% versus 13.6%) and extrapancreatic infections (21.6% versus 23.9%)<br />

and clinical outcome.<br />

• Summary: Meropenem is as effective as imipenem in preventing septic<br />

complications of patients with severe acute pancreatitis.<br />

In order to determine the in vitro effects of combinations of meropenem, cefepime,<br />

and piperacillin-tazobactam with ciprofloxacin against multidrug-resistant Ps.<br />

aeruginosa:<br />

• The results show that both the combination of two β-lactams with<br />

ciprofloxacin and three β-lactams with ciprofloxacin against 4 of 5<br />

multidrug-resistant P. aeruginosa strains has no effect in vitro.<br />

• The combination of one β-lactam (meropenem, cefepime, and piperacillintazobactam)<br />

with ciprofloxacin has a synergistic effect against one strain of<br />

multidrug-resistant P. aeruginosa that is ciprofloxacin susceptible and<br />

resistant to meropenem, cefepime, and piperacillin-tazobactam.<br />

• None of the combinations had an antagonistic effect against these<br />

multidrug-resistant strains.<br />

Meropenem vs. n=54<br />

combination pediatric<br />

therapy with cancer<br />

ceftazidime plus patients<br />

amikacin for<br />

pediatric cancer<br />

patients with<br />

neutropenia 46<br />

Results of the<br />

Meropenem<br />

Yearly<br />

Susceptibility<br />

Test Information<br />

Collection<br />

(MYSTIC)<br />

Program 47<br />

n=2,874<br />

strains<br />

processed<br />

from 15 U.S.<br />

medical<br />

centers<br />

- In evaluating the febrile episodes in pediatric cancer patients treated with meropenem<br />

or ceftazidime plus amikacin as empirical therapy:<br />

• The success rate with unmodified therapy was not significantly different<br />

between the meropenem group (72%) and the ceftazidime-plus-amikacin<br />

group (57%).<br />

• The incidence of side effects was similar between the two groups and these<br />

side effects were reversible.<br />

• The clinical response rates in subgroups of documented infection and<br />

unexplained fever did not significantly differ between the two treatment<br />

groups.<br />

• Meropenem was significantly more effective than ceftazidime plus<br />

amikacin in children at high risks of developing severe infection who had<br />

profound neutropenia (absolute neutrophil count [ANC] < 100/mm 3 ),<br />

prolonged neutropenia (ANC < 500/mm 3 lasting for > 10 days), or<br />

clinically deteriorating shock (p=0.045).<br />

• Summary: As an empirical treatment, meropenem seems to be as effective<br />

and safe as ceftazidime plus amikacin for febrile episodes in children with<br />

cancer and neutropenia. Meropenem is more effective for pediatric cancer<br />

patients at the high risk of severe infection.<br />

- To monitor in vitro activity of meropenem and other broad-spectrum antimicrobial<br />

agents, in institutions where meropenem is the primary therapeutic carbapenem:<br />

• The meropenem MIC(90) values were 0.03mg/l for Citrobacter spp.,<br />

Escherichia coli and Klebsiella spp.; 0.06mg/l for Proteus mirabilis and<br />

Serratia spp. and 0.12 mg/l for Enterobacter spp. This potency was 8 to 16-<br />

fold greater than that of imipenem. The meropenem spectrum of activity<br />

versus the Enterobacteriaceae was the broadest of all tested antimicrobial<br />

agents.<br />

• Only piperacillin/tazobactam (MIC(9), 64mg/l) and tobramycin (MIC(90),<br />

4mg/l) were active against more than 90.0% of Pseudomonas aeruginosa at<br />

the NCCLS susceptible breakpoint, and the carbapenems were the most<br />

active compounds against Acinetobacter spp.<br />

• However, Acinetobacter spp. isolates were resistant to all of the<br />

antimicrobial agents tested and the molecular typing results suggested that<br />

they were epidemiologically related.<br />

• Only ciprofloxacin and ceftazidime had significantly reduced activity<br />

against oxacillin-susceptible staphylococci (87.9-92.6% susceptible).<br />

• Summary: These 2001 US MYSTIC Program results demonstrated no<br />

significant decline in carbapenem susceptibility rates compared with the<br />

previously monitored years. Most apparent were the decreasing<br />

susceptibility rates for ciprofloxacin and ceftazidime against staphylococci.<br />

148


Randomized,<br />

multicenter,<br />

open-label study<br />

of meropenem vs.<br />

imipenem/<br />

cilastatin for<br />

serious intensive<br />

care unit<br />

infections 48 n=178 - In comparing the efficacy and tolerability of meropenem and imipenem/cilastatin as<br />

empirical monotherapy in intensive care unit (ICU) patients with serious bacterial<br />

infections:<br />

• The overall satisfactory clinical response rate at the end of randomized<br />

treatment was 77.0% (67/87) with meropenem and 68.1% (62/91) with<br />

imipenem/cilastatin (difference 8.9%; 95% confidence interval -4.2% to<br />

21.9%; p = 0.185).<br />

• The two drugs produced similar satisfactory clinical response rates against<br />

lower respiratory infections: 68.3% (41/60) with meropenem versus 68.6%<br />

(35/51) with imipenem/cilastatin.<br />

• Meropenem appeared to be slightly more effective against intra-abdominal<br />

infections: 95.5% (21/22) versus 76.7% (23/30), respectively.<br />

• All five meropenem recipients with sepsis had a satisfactory clinical<br />

response, compared to 40.0% (4/10) of those who received<br />

imipenem/cilastatin.<br />

• The overall satisfactory bacteriologic response rate was 67.1% (49/73) with<br />

meropenem and 60.3% (44/73) with imipenem/cilastatin (difference 6.9%;<br />

95% confidence interval -8.7% to 22.4%; p = 0.389).<br />

• No incidences of drug-related nausea and vomiting were reported, but one<br />

probable drug-related seizure occurred in the imipenem/cilastatin group.<br />

• Summary: Meropenem is at least as efficacious (clinically and<br />

bacteriologically) as imipenem/cilastatin for the empirical monotherapy of<br />

serious bacterial infections in ICU patients, and it can therefore be<br />

The Meropenem<br />

Yearly<br />

Susceptibility<br />

Test Information<br />

Collection<br />

(MYSTIC)<br />

Program 49<br />

n=2,848<br />

isolates<br />

Antimicrobial activity<br />

of 11 broad-spectrum<br />

agents was assessed<br />

considered a useful option in this setting.<br />

In evaluating the spectrum of activity and potency of meropenem within medical<br />

centers where carbapenems are used for the treatment of serious infections:<br />

• The minimum inhibitory concentration (MIC) results demonstrate the<br />

continued high potency of meropenem against all monitored pathogens.<br />

• Against all gram-negative bacilli tested, the overall rank order of<br />

susceptibility was meropenem (96.3%) > imipenem (95.6%) > cefepime<br />

(93.7%) > tobramycin (91.9%) > piperacillin/tazobactam (90.2%) ><br />

ceftazidime (90.1%) > gentamicin (89.6%) > levofloxacin (82.8%) ><br />

ciprofloxacin (82.5%) > aztreonam (81.8%) > ceftriaxone (72.3%).<br />

Meropenem in n=62<br />

cystic fibrosis patients with<br />

patients with cystic<br />

resistant Ps. fibrosis<br />

aeruginosa 50<br />

*Product not approved in the United States.<br />

Meropenem 2g TID for<br />

14 days<br />

To assess the efficacy and safety of meropenem, administered on a compassionate<br />

basis in cystic fibrosis patients chronically infected with Ps. aeruginosa:<br />

• During treatment for P. aeruginosa the mean increase in pulmonary<br />

function (as a percentage of the predictive values) was 5.6% for FEV1<br />

(forced expiratory volume in the first second) and 8.6% for FVC (forced<br />

vital capacity).<br />

• C-reactive protein and erythrocyte sedimentation rate (ESR) and leukocyte<br />

count decreased significantly.<br />

• In courses administered for chronic infection with B. cepacia the post<br />

treatment FEV1 and FVC values were higher than the pre-treatment values,<br />

and all the inflammatory parameters decreased.<br />

• The geometric means of minimal inhibitory concentration (MICs)<br />

(microg/mL) for P. aeruginosa (B. cepacia) were: tobramycin 6 (59),<br />

ciprofloxacin 1.2 (9.7), piperacillin 49 (16.3), ceftazidime 26 (23),<br />

aztreonam 26 (35), imipenem 6.4 (not determined) and meropenem 5.1<br />

(4.8).<br />

• No statistically significant increase in the MICs of meropenem for either<br />

pathogen occurred during therapy.<br />

• Of the 124 courses, 115 were tolerated without any clinical complaint. The<br />

following side effects were observed: nausea (0.8%), itching (4%), rash<br />

(3.2%), and drug fever (1.6%).<br />

• Summary: Meropenem proved to be a valuable drug in the treatment of CF<br />

patients with chronic pulmonary infection with multiresistant P.<br />

aeruginosa and B. cepacia and with hypersensitivity reactions to other<br />

beta-lactam drugs.<br />

149


Additional Evidence<br />

Dose Simplification: The drugs in this class are for serious bacterial infections. Most<br />

frequently, the IV therapies are given during hospitalization or via nursing care within extended<br />

care facilities. There are no alternative dosing options for most drugs in this class. Additionally,<br />

the only oral medication in this class, loracarbef, is given twice daily. Ertapenem (QD) has a less<br />

frequent dosing regimen compared to meropenem (Q8). A literature search did not reveal data<br />

that has looked at the impact of a less frequent dosing regimen and impact on the outcome of the<br />

infectious disease.<br />

Stable Therapy: Not applicable. Some studies presented above have addressed resistance with<br />

drugs in this class.<br />

Impact on Physician Visits: Due to the nature of the use of the drugs in this class, and to their<br />

indications, no published studies have evaluated the impact of use of these drugs on physician<br />

visits. A search of Medline and Ovid did not reveal data pertinent to this topic.<br />

IX.<br />

Conclusions<br />

The drugs in this class are indicated for use in conditions from septicemia, to intra-abdominal<br />

infections and meningitis. These drugs would not routinely be used as first-line therapies on an<br />

outpatient basis. Aztreonam may be an alternative to the use of aminoglycosides, especially for<br />

use against atypicals and in patients with allergies to other antibiotics with gram-negative<br />

coverage. Clinical studies have shown ertapenem to be comparable to piperacillin/tazobactam and<br />

ceftriaxone (depending on the use). Ertapenem does have once-daily dosing, where meropenem<br />

and imipenem/cilastatin are dosed more frequently. The most common use for cefotetan and<br />

cefoxitin is in surgery prophylaxis and abdominal or gynecological infections. Loracarbef has a<br />

spectrum of coverage similar to cefaclor, a 2 nd generation cephalosporin, which is available<br />

generically. 51 Studies have shown loracarbef to have similar efficacy compared to doxycycline,<br />

norfloxacin, clarithromycin, and cefdinir. Treatment with meropenem is an option for empiric<br />

therapy in polymicrobic infections or with multi-resistant organisms, and studies have shown<br />

meropenem to be comparable or as effective as imipenem/cilastatin.<br />

In most cases, the drugs in this class are given to hospitalized patients, due to the serious nature of<br />

the indications and their injectable availability (with the exception of loracarbef). It is not,<br />

however, out of the question that a recipient could be discharged from a hospital institution to an<br />

extended-care facility or home, where antibiotic therapy could be continued for the remainder of<br />

the treatment course. In the event this does occur, the patient should be allowed to continue<br />

therapy with the same antibiotic.<br />

Although there may be some clinical advantage to drugs in this class in special<br />

needs/circumstances, this would not be considered applicable in general use. Therefore, all brand<br />

products within the class reviewed are comparable to each other and to the generics in the class<br />

and offer no significant clinical advantage over other alternatives in general use.<br />

X. Recommendations<br />

No brand single entity miscellaneous β-lactam antibiotic is recommended for preferred status.<br />

150


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Miscellaneous β-Lactam Antibiotics<br />

Combination Agents<br />

AHFS 081207<br />

I. Overview<br />

Imipenem is a beta-lactam antibiotic derived from thienamycin and was the first drug to be<br />

classified as a carbapenem antibiotic. It is the only combination miscellaneous β-lactam drug<br />

available. Cilastatin is added to imipenem as an inhibitor of dehydropeptidase-1, an enzyme<br />

found in the renal tubule border that metabolizes imipenem. 6 Without cilastatin, imipenem is<br />

rapidly metabolized and causes toxicity to the proximal tubule. Cilastatin itself has no<br />

antibacterial activity. Imipenem possesses several traits that make it an effective antibiotic<br />

including: a) more efficient penetration through the bacterial cell wall, b) resistance to bacterial<br />

enzymes, and c) affinity for all bacterial PBPs. Imipenem has a broader spectrum of activity than<br />

do many other β-lactam antibiotics. Clinically, the combination of imipenem-cilastatin is used to<br />

treat severe or resistant infections, especially those that are nosocomial in origin. The FDA<br />

approved imipenem-cilastatin in November 1985. There is no generic formulation available for<br />

imipenem/cilastatin. This review encompasses all dosage forms and strengths.<br />

Table 1. Combination Misc. B-Lactam Antibiotics in this <strong>Review</strong><br />

Generic Name β-Lactam Classification Formulation Example Brand Name<br />

Imipenem/cilastatin Carbapenem Injection (IV and IM) Primaxin<br />

Table 2. Comparison of Bacterial Coverage of Imipenem/cilastatin 5<br />

Drug<br />

Spectrum<br />

Imipenem Cilastatin is added as an inhibitor of dehydropeptidase-1, an enzyme found in the<br />

renal tubule border that metabolizes imipenem. Without cilastatin, imipenem is<br />

rapidly metabolized and causes toxicity to the proximal tubule. Cilastatin itself has<br />

no antibacterial activity. Imipenem possesses several traits that make it an effective<br />

antibiotic including: a) more efficient penetration through the bacterial cell wall, b)<br />

resistance to bacterial enzymes, and c) affinity for all bacterial penicillin binding<br />

proteins (PBPs). Imipenem has a broader spectrum of activity than do many other<br />

β-lactam antibiotics. Clinically, the combination of imipenem-cilastatin is used to<br />

treat severe or resistant infections, especially those that are nosocomial in origin.<br />

Imipenem is active against a wide variety of organisms. The gram-positive<br />

organisms susceptible to imipenem include Enterococcus faecalis, group A, C, and<br />

G streptococci, Streptococcus pneumoniae, and group D enterococci as well as the<br />

penicillinase- and nonpenicillinase-producing staphylococci and Listeria<br />

monocytogenes. In vitro, imipenem is inactive against Enterococcus faecium.<br />

Activity against methicillin-resistant strains of staphylococci is variable but, it is<br />

recommended that these strains be reported as resistant to imipenem. Imipenem's<br />

gram-negative spectrum includes N. meningitidis, N. gonorrhoea, H. influenzae,<br />

Branhamella catarrhalis, Acinetobacter, Aeromonas hydrophila, Campylobacter<br />

jejuni, Pasteurella multocida, and most Enterobactericeae (E.coli, Klebsiella,<br />

Citrobacter, Eneterobacter, Morganella, Proteus, Providencia, Serratia,<br />

Salmonella, Shigella, and Yersinia). Imipenem has exceptional stability against β-<br />

lactamases. It is highly active against Enterobacteriaceae that are resistant to thirdgeneration<br />

cephalosporins. Also, imipenem exhibits good activity against<br />

Pseudomonas aeruginosa, similar to that of ceftazidime. However, imipenem is<br />

inactive in vitro against Xanthomonas (Pseudomonas) maltophillia and some strains<br />

of P. cepacia. The anaerobic spectrum of imipenem includes Bacteriodes fragilis,<br />

Clostridium perfringens, Clostridium tetani, and Peptococcus and<br />

Peptostreptococcus species.<br />

151


II.<br />

Indications of the Combination Miscellaneous β-Lactam Antibiotics<br />

Table 3. FDA-Approved Indications for the Combination Miscellaneous β-Lactams 5-8<br />

Drug UTI Lower<br />

RTI<br />

Gynecologic<br />

Infections<br />

Septicemia<br />

Imipenem/<br />

Cilastatin<br />

✔<br />

Complicated<br />

and<br />

uncomplicated<br />

Intraabdominal<br />

Infections<br />

* May use IM route for non life-threatening infections.<br />

Bone and<br />

Joint<br />

Infections<br />

Skin and<br />

Skin<br />

Structure<br />

Infections<br />

Endocarditis<br />

Polymicrobic<br />

Infections<br />

✔* ✔* ✔* ✔ ✔ ✔* ✔ ✔<br />

III.<br />

Pharmacokinetic Parameters<br />

Table 4. Pharmacokinetic Parameters of the Combination Miscellaneous β-Lactams 5-8<br />

Drug Mechanism of Action Bioavailability Protein<br />

Binding<br />

Metabolism Active<br />

Metabolites<br />

Imipenem/<br />

cilastatin<br />

Imipenem is mainly<br />

bactericidal. It inhibits the<br />

third and final stage of<br />

bacterial cell wall<br />

synthesis by preferentially<br />

binding to specific<br />

penicillin-binding proteins<br />

(PBPs) that are located<br />

inside the bacterial cell<br />

wall.<br />

Peak plasma<br />

levels occur in<br />

20 minutes<br />

following an<br />

IV dose<br />

20%<br />

imipenem<br />

and 40%<br />

cilastatin<br />

Cilastatin is<br />

metabolized<br />

by the<br />

kidneys<br />

Elimination<br />

- 70% of a<br />

dose of<br />

imipenem<br />

(when<br />

given with<br />

cilastatin)<br />

is excreted<br />

in urine,<br />

within 10<br />

hours<br />

Half-<br />

Life<br />

60<br />

minutes<br />

in<br />

normal<br />

renal<br />

function<br />

IV.<br />

Drug Interactions<br />

Table 5 lists the most significant drug drug-interactions (Level 1 and 2) for the drugs indexed by<br />

Drug Interactions Facts. 9<br />

Table 5. Drug Interactions of the Combination Miscellaneous β-Lactams 9<br />

Drug Significance Interaction Mechanism<br />

Imipenem/cilastatin Level 2 Imipenem/cilastatin and<br />

cyclosporine<br />

This interaction may be the result of<br />

additive or synergistic toxicity. The<br />

CNS side effects of both agents may be<br />

increased.<br />

Other reported side effects for imipenem/cilastatin 8 :<br />

Imipenem-Cilastatin Drug Interactions<br />

Precipitant Drug Object Drug * Description<br />

Imipenem-<br />

Cilastatin<br />

Ganciclovir Generalized seizures have occurred with coadministration. Do not use concomitantly.<br />

Probenecid Imipenem Coadministration results in only minimal increases in imipenem levels and half-life; do<br />

not give probenecid concurrently.<br />

*<br />

= Object drug increased.<br />

152


V. Adverse Events of the Combination Miscellaneous β-Lactam Antibiotics<br />

Table 6. Common Adverse Events (%) Reported for the Combination Miscellaneous β-Lactams 6-8<br />

Adverse Event<br />

Body as a Whole<br />

Malaise<br />

Cardiovascular<br />

Vasodilation<br />

Hypotension<br />

Hypertension<br />

Transient ECG Changes<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Epigastric distress<br />

Appetite decrease<br />

Constipation<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

Insomnia<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Erythema multiforme<br />

Hematologic<br />

Neutropenia<br />

Thrombocytopenia<br />

Leukopenia<br />

Eosinophilia<br />

Anemia<br />

Thrombocytosis<br />

Bleeding events<br />

Imipenem/Cilastatin<br />

0.4<br />


VI.<br />

Dosage and Administration for the Combination Miscellaneous β-Lactam<br />

Antibiotics<br />

Table 7. Dosing for the Combination Misc. B-Lactam Antibiotics 5-8<br />

Drug Availability Dose/Frequency/Duration<br />

IV: 250mg and 500mg<br />

powder for injection,<br />

also available in Add-<br />

Vantage and Monovial<br />

vials;<br />

IM: 500mg and 750mg<br />

powder for injection<br />

Imipenem/<br />

cilastatin<br />

IV :<br />

Give a 125, 250, or 500mg dose by IV infusion over 20 to 30 minutes.<br />

Infuse a 750mg or 1g dose over 40 to 60 minutes. If nausea develops,<br />

slow the infusion rate.<br />

Because of high antimicrobial activity, do not exceed 50mg/kg/day or<br />

4g/day, whichever is lower. There is no evidence that higher doses<br />

provide greater efficacy.<br />

Type or severity<br />

of infection<br />

Imipenem-Cilastatin IV Dosing Schedule for Adults<br />

with Normal Renal Function<br />

Fully susceptible<br />

organisms 1<br />

Total<br />

daily<br />

dose<br />

Moderately susceptible<br />

organisms, primarily<br />

some strains of<br />

P. aeruginosa<br />

Total<br />

daily<br />

dose<br />

Mild 250mg q 6 hr 1g 500mg q 6 hr 2g<br />

Moderate<br />

Severe,<br />

life-threatening<br />

Uncomplicated<br />

UTI<br />

500mg q 8 hr<br />

or 500mg q 6 hr<br />

1.5 or<br />

2g<br />

500mg q 6 hr<br />

or 1g q 8 hr<br />

500mg q 6 hr 2g 1g q 8 hr<br />

or 1g q 6 hr<br />

2 or<br />

3g<br />

3 or<br />

4g<br />

250mg q 6 hr 1g 250mg q 6 hr 1g<br />

Complicated 500mg q 6 hr 2g 500mg q 6 hr 2g<br />

UTI<br />

1 Including gram-positive and -negative aerobes and anaerobes<br />

IM :<br />

Total daily IM dosages > 1500mg/day are not recommended.<br />

Duration of therapy depends on the type and severity of the infection.<br />

Generally, continue for ≥2 days after signs and symptoms of infection<br />

have resolved. Safety and efficacy of treatment > 14 days have not been<br />

established.<br />

Administer by deep IM injection into a large muscle mass (such as the<br />

gluteal muscles or lateral part of the thigh) with a 21-gauge 2-inch needle.<br />

Aspiration is necessary to avoid inadvertent injection into a blood vessel.<br />

Imipenem-Cilastatin IM Dosage Guidelines in Adults<br />

Type/Location of<br />

infection Severity Dosage regimen<br />

Lower respiratory tract<br />

Skin and skin structure<br />

Gynecologic<br />

Mild/Moderate 500 or 750mg q12 hr<br />

depending<br />

on the severity of infection<br />

Intra-abdominal<br />

Mild/Moderate 750mg q 12 hr<br />

154


Children:<br />

In mild-to-moderate infections the recommended dose is 10 to<br />

15mg/kg every 6 hours.<br />

Pediatric Dosing Guidelines (≥3 months old)<br />

15 to 25mg/kg/dose every 6 hours<br />

≥3 months old (non-CNS infections)<br />

Maximum daily dose for fully susceptible organisms is 2g/day, and for<br />

infections with moderately susceptible organisms (primarily some strains<br />

of P. aeruginosa)is 4g/day (based on adult studies).<br />

Higher doses (≤90mg/kg/day in older children) have been used in<br />

cystic fibrosis patients.<br />

Pediatric Dosing Guidelines(≤3 months old)<br />

≤3 months old (weighing ≥1500g; non-CNS infections)<br />

< 1 week old: 25mg/kg every 12 hours<br />

1 to 4 weeks old: 25mg/kg every 8 hours<br />

4 weeks to 3 months old: 25mg/kg every 6 hours<br />

Give doses ≤500mg by IV infusion over 15 to 30 minutes. Give doses<br />

>500mg by IV infusion over 40 to 60 minutes.<br />

Imipenem-cilastatin IV is not recommended in pediatric patients with<br />

CNS infections because of the risk of seizures and in pediatric patients<br />


VII.<br />

Comparative Efficacy<br />

Imipenem/cilastatin can be compared with the other carbapenems, ertapenem and meropenem.<br />

Imipenem has stronger coverage for Enterococcus faecalis, Enterococcus faecium, and Y.<br />

enterocolitica as compared to ertapenem and meropenem. 51 Multiple studies presented in the<br />

single-entity review comparing meropenem and imipenem/cilastatin showed no statistically<br />

significant differences in clinical outcome between the two agents. Little data is available directly<br />

comparing imipenem/cilastatin with ertapenem. Table 9 describes additional comparative data for<br />

the carbapenems.<br />

Table 9. Additional Outcomes Evidence for the Combination Miscellaneous β-Lactams<br />

Study Sample Treatment/Duration Results<br />

Randomized study of<br />

carbapenems vs.<br />

standard therapy for<br />

CAP 52 n=204 Meropenem 500mg<br />

TID,<br />

imipenem/cilastatin<br />

500mg TID,<br />

In evaluating the carbapenems versus standard therapy for elderly hospitalized<br />

patients with community-acquired pneumonia:<br />

• A satisfactory clinical, bacteriological response was achieved<br />

respectively in 86.5% in meropenem; 86.3% in imipenem/cilastatin;<br />

clarithromycin 500mg +<br />

ceftriaxone 1g BID,<br />

clarithromycin 500mg +<br />

amikacin 250mg BID<br />

69% in ceftriaxone + clarithromycin and in 85.7% in clarithromycin +<br />

amikacin.<br />

• This study shows that treatment with either meropenem or imipenem is<br />

as efficacious as conventional therapy in the treatment of community<br />

Empiric carbapenem<br />

monotherapy in<br />

pediatric bone<br />

marrow recipients 53 n=32 Prospective analysis of<br />

meropenem patients and<br />

retrospective analysis of<br />

imipenem/cilastatin<br />

patients<br />

acquired pneumonia (CAP).<br />

To determine which carbapenem is the preferable empiric antibiotic<br />

monotherapy in pre-engrafted pediatric bone marrow transplant in terms of<br />

patient tolerance and therapeutic efficacy:<br />

• No differences were found in the number of culture proven or<br />

clinically suspected breakthrough bacterial infections or the need for<br />

concurrent additional antibiotics between the groups.<br />

• Our analysis found that patients who received meropenem<br />

experienced significantly less vomiting than those in the<br />

imipenem/cilastatin cohort. The statistical and clinical differences<br />

in the number of vomiting episodes between these groups impacted<br />

other aspects of patient care, antiemetic use, and TPN duration.<br />

Randomized,<br />

controlled trial of<br />

meropenem and<br />

imipenem/cilastatin<br />

for acute bacterial<br />

infections 54 n=112 Meropenem 500mg<br />

Q12 (or 1g Q12) vs.<br />

imipenem/cilastatin<br />

500mg Q12 (or 1g Q12)<br />

Randomized,<br />

n=144<br />

multicenter trial of patients<br />

meropenem vs. age 18-<br />

imipenem/cilastatin 94 years<br />

for communityacquired<br />

pneumonia 55<br />

Meropenem 1.5g daily<br />

vs. imipenem/cilastatin<br />

2g daily; the mean<br />

duration of treatment<br />

was 10 days for<br />

meropenem and 9.7<br />

days for<br />

imipenem/cilastatin<br />

In comparing the effectiveness of meropenem with imipenem for acute<br />

bacterial infections:<br />

• 42 of the 55 cases receiving meropenem and 41 of the 57 cases<br />

receiving imipenem/cilastatin were assessable for clinical efficacy.<br />

• The overall efficacy rate was 88.1% (37/42) for the meropenem<br />

group and 85.4% (35/41) for the imipenem/cilastatin group, whereas<br />

the bacterial eradication rate was 81.1% (30/37) and 84.2% (32/38),<br />

respectively. 47 (69.1%) of 68 strains isolated from patients<br />

produced beta-lactamase.<br />

• Adverse drug reaction was evaluated in 44 cases of the meropenem<br />

group and 41 cases of the imipenem/cilastatin group. The adverse<br />

drug reaction rate was 13.6% (6/44) and 12.2% (5/41), respectively.<br />

• The results showed that there were no statistical differences between<br />

these two groups (p > 0.05).<br />

In comparing the efficacy and safety of meropenem and imipenem/cilastatin in<br />

the hospital treatment of community-acquired pneumonia:<br />

• At the end of therapy, cure or improvement in signs and symptoms<br />

as a satisfactory clinical response was observed in 57 of 64 (89.1%)<br />

meropenem-treated patients and in 60 of 66 (90.9%)<br />

imipenem/cilastatin patients.<br />

• In patients who were followed up for weeks 2-4, the response was<br />

satisfactory (100%) for both treatments.<br />

• A satisfactory bacteriological response, defined as either presumed<br />

or confirmed eradication of all pathogens, was found in eight<br />

patients who had received meropenem and in 14 patients who had<br />

received imipenem/cilastatin. Response was considered satisfactory<br />

in 100% of the meropenem group and in 92.9% of the<br />

imipenem/cilastatin group and at follow-up, it was 100% for both<br />

treatments.<br />

156


• Drug-related adverse events were reported in three (4.2%)<br />

meropenem-treated patients and in eight (11.0%)<br />

imipenem/cilastatin-treated patients.<br />

• Summary: The clinical and bacteriological efficacy and tolerability<br />

of meropenem are similar to that of imipenem/cilastatin in the<br />

hospital treatment of community-acquired pneumonia.<br />

Additional Evidence<br />

Dose Simplification: Imipenem/cilastatin is a potent antibiotic for serious bacterial infections.<br />

This medication would most likely be given during hospitalization or potentially through nursing<br />

care within an extended care facility. Meropenem (Q8) and ertapenem (QD) have less frequent<br />

dosing regimens as compared to imipenem, however, clinical data is not available that has<br />

evaluated the dosing frequency difference and outcome of the infectious disease.<br />

Stable Therapy: Not applicable.<br />

Impact on Physician Visits: A search of Medline and Ovid did not reveal data pertinent to<br />

medical resource utilization or the duration of hospitalization.<br />

VIII. Conclusion<br />

Clinical data suggests imipenem/cilastatin and meropenem are similar or comparable in efficacy<br />

and safety. Although ertapenem has a comparable spectrum of coverage, less clinical comparative<br />

data is available for ertapenem.<br />

Imipenem, like the single entity miscellaneous β-Lactams, would not routinely be used as first-line<br />

therapy on an outpatient basis. Imipenem/cilastatin is an agent that would not be considered<br />

applicable to general use in the population. Imipenem’s role is in the treatment of polymicrobic<br />

infections and other serious infections usually requiring hospitalization. Should therapy need to<br />

be continued beyond the hospital stay, imipenem/cilastatin should be available for the remainder<br />

of the course of therapy.<br />

Again, there may be some clinical advantage to the drugs within this combination class review in<br />

special needs/circumstances, but use of these agents would not be considered applicable in general<br />

use. Therefore, all brand products within the class reviewed are comparable to each other and to<br />

the generics in the class and offer no significant clinical advantage over other alternatives in<br />

general use.<br />

IX.<br />

Recommendation<br />

No brand combination miscellaneous β-lactam antibiotic is recommended for preferred status.<br />

157


References<br />

1. Asbel LE, Levison ME. Cephalosporins, carbapenems, and monobactams. Infect Dis Clin North<br />

Am 2000 Jun;14(2):435-47.<br />

2. Lizasoain M, Noriega AR. Tolerance and safety of carbapenems: the use of meropenem.<br />

Enferm Infec Microbiol Clin 1997 Sep 15 Suppl 1:73-7.<br />

3. The ASCAP Panel Consensus Report, 2002. Community-acquired pneumonia (CAP) year 2002,<br />

antibiotic selection and patient management update. Available at: www.clinicalconsensusreports.com.<br />

Accessed September 15, 2004.<br />

4. Mandell LA, Bartlett JG, Dowell SF, et al. Update of practice guidelines for the management of<br />

community-acquired pneumonia. Clin Inf Dis 2003 Dec 1;37:1405-33.<br />

5. Clinical Pharmacology, 2004. Available on-line at: www.cp.gsm.com. Accessed September 15,<br />

2004.<br />

6. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58,<br />

2004. Thomson PDR. Montvale, NJ. 2004.<br />

7. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

8. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

9. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

10. Briggs GG, Freeman RK, Yaffe SJ. Drugs in pregnancy and lactation. A reference guide to fetal<br />

and neonatal risk. Sixth Edition. Lippincott, Williams, & Wilkins, Philadelphia, 2002.<br />

11. Lorabid ® [package insert]. Bristol, TN: Monarch Pharmaceuticals; September 2002.<br />

12. Schatz BS, Karavokiros KT, Taeubel MA, et al. Comparison of cefprozil, cefpodoxime,<br />

loracarbef, cefixime, and ceftibuten. Ann Pharmacother 1996 Mar;30(3):258-68.<br />

13. Oie S, Uematsu T, Sawa A, et al. In vitro effects of combinations of antipseudomonal agents<br />

against seven strains of multidrug-resistant Pseudomonas aeruginosa. J Antimicrob Chemother 2003<br />

Dec;52(6):911-4.<br />

14. Karlowsky JA, Draghi DC, Jones ME, et al. Surveillance for antimicrobial susceptibility among<br />

clinical isolates pf Pseudomonas aeruginosa and Acinetobacter baumannii from hospitalized patients in<br />

the United States, 1998 to 2001. Antimicro Agents Chemother 2003 May;47(5):1681-8.<br />

15. Rhomberg PR, Jones RN; MYSTIC Program (USA) Study Group. Antimicrobial spectrum of<br />

activity for meropenem and nine broad spectrum antimicrobials: report from the MYSTIC Program.<br />

Diagn Microbiol Infect Dis 2003 Sep;47(1):365-72.<br />

16. Jones RN, Biedenbach DJ, Gales AC. Sustained activity and spectrum of selected extendedspectrum<br />

B-lactams (carbapenems and cefepime) against Enterobacter spp. ESBL-producing<br />

Klebsiella spp.: report from the SENTRY antimicrobial surveillance program (USA, 1997-2000).<br />

17. Jones RN, Kirby JT, Beach ML, et al. Geographic variations in activity of broad-spectrum betalactams<br />

against Pseudomonas aeruginosa: summary of the worldwide SENTRY Antimicrobial<br />

Surveillance Program (1997-2000). Diagn Microbiol Infect Dis 2002 Jul;43(3):239-43.<br />

18. Holzheimer RG, Dralle H. Antibiotic therapy in intra-abdominal infections—a review on<br />

randomized clinical trials. Eur J Med Res 2001 Jul 30;6(7):277-91.<br />

19. Sader HS, Huynh HK, Jones RN. Contemporary in vitro synergy rates for aztreonam combined<br />

with newer fluoroquinolones and beta-lactams tested against gram-negative bacilli. Diagn Microbiol<br />

Infect Dis 2003 Nov;47(3):547-50.<br />

20. Boucher BA. Role of aztreonam in the treatment of nosocomial pneumonia in the critically ill<br />

surgical patient. Am J Surg 2000 Feb 1;179(2 Suppl 1):45-50.<br />

21. Fleming DR, Zeigler c, Baize T, et al. Cefepime versus ticarcillin and clavulanate potassium and<br />

aztreonam for febrile neutropenia therapy in high-dose chemotherapy patients. Am J Clin Oncol 2003<br />

Jun;26(3):285-8.<br />

22. Khan MT, Shah SH. Comparison of aztreonam against other antibiotics used in urinary tract<br />

infections. J Ayub Med Coll Abbottabad 2001 Oct-Dec;13(4):22-4.<br />

23. Ballow CH, Wels PB, Welage LS, et al. A double-blind, randomized comparison of aztreonam<br />

plus clindamycin with tobramycin plus clindamycin in abdominal infections. Am J Ther 1995<br />

Jun;2(6):373-377.<br />

24. Doganov N, Shtereva K, Dimitrov R. A comparative study of the efficacy of antibiotic<br />

prophylaxis with cephalosporins in operative gynecology.<br />

25. Gugino L, Cimino M, Wactawski-Wende J. Single-dose antibiotic prophylaxis during cesarean<br />

section. Prim Care Update Ob Gyns 1998 Jul 1;5(4):147-148.<br />

158


26. Gesser RM, McCarroll KA, Woods GL. Evaluation of outpatient treatment with ertapenem in a<br />

double-blind controlled clinical trial of complicated skin/skin structure infections. I Infect 2004<br />

Jan;48(1):32-8.<br />

27. Gesser RM, McCarroll KA, Woods GL. Efficacy of ertapenem against methicillin-susceptible<br />

Staphylococcus aureus in complicated skin/skin structure infections: results of a double-blind<br />

clinical trial versus piperacillin-tazobactam. Int J Antimicrob Agents 2004 Mar;23(3):235-9.<br />

28. Wells WG, Woods GL, Jiang Q, et al. Treatment of complicated urinary tract infection in adults:<br />

combined analysis of two randomized, double-blind, multicenter trials comparing ertapenem and<br />

ceftriaxone followed by appropriate oral therapy. J Antimicrob Chemother 2004 Jun;53 Suppl<br />

2:ii67-74.<br />

29. Solomkin J, Teppler H, Graham DR, et al. Treatment of polymicrobial infections: post hoc<br />

analysis of three trials comparing ertapenem and piperacillin-tazobactam. J Antimicrob<br />

Chemother 2004 Jun;53 Suppl 2:ii51-7.<br />

30. Alhambra A, Cuadros JA, Cacho J, et al. In vitro susceptibility of recent antibiotic-resistant<br />

urinary pathogens to ertapenem and 12 other antibiotics. J Antimicrob Chemother 2004<br />

Jun;53(6):1090-4.<br />

31. Vetter N, Cambronero-Hernandez E, Rohlf J, et al. A prospective, randomized, double-blind<br />

multicenter comparison of parenteral ertapenem and ceftriaxone for the treatment of hospitalized<br />

adults with community-acquired pneumonia. Clin Ther 2002 Nov;24(11):1770-85.<br />

32. Unnamed authors. Loracarbef versus doxycycline in the treatment of acute bacterial maxillary<br />

sinusitis. Scandinavian Study Group. J Antimicrob Chemother 1993 Jun;31(6):949-61.<br />

33. Hyslop DL, Bischoff W. Loracarbef (LY163892) versus cefaclor and norfloxacin in the treatment<br />

of uncomplicated pyelonephritis. Am J Med 1992 Jun 22;92(6A):86S-94S.<br />

34. Muller O, Spirer Z, Wettich K. Loracarbef versus penicillin V in the treatment of streptococcal<br />

pharyngitis and tonsillitis. Infection 1992 Sep-Oct;20(5):301-8.<br />

35. Hanfling MJ, Hausinger SA, Squires J. Loracarbef vs. cefaclor in pediatric skin and skin structure<br />

infections. Pediatr Infect Dis J 1992 Aug;11(8 Suppl):S27-30.<br />

36. Kays MB, Wood KK, Miles DO. In vitro activity and pharmacodynamics of oral beta-lactam<br />

antibiotics against Streptococcus pneumoniae from southeast Missouri. <strong>Pharmacotherapy</strong> 1999<br />

Nov;19(11):1308-14.<br />

37. Gooch WM 3 rd , Adelglass J, Kelsey DK, et al. Loracarbef versus clarithromycin in children with<br />

acute otitis media with effusion. Clin Ther 1999 Apr;21(4):711-22.<br />

38. Paster RZ, McAdoo MA, Keyserling CH, et al. A comparison of a five-day regimen of cefdinir<br />

with a seven-day regimen of loracarbef for the treatment of acute exacerbations of chronic<br />

bronchitis. Int J Clin Pract 2000 Jun;54(5):293-9.<br />

39. Demers DM, Chan DS, Bass JW. Antimicrobial drug suspensions: a blinded comparison of taste<br />

of twelve common pediatric drugs including cefixime, cefpodoxime, cefprozil and loracarbef.<br />

Pediatr Infect Dis J 1994 Feb;13(2):87-9.<br />

40. Greenhalgh JM, Edwards JR. A comparative study of the in vitro activity of meropenem and<br />

representatives of the major classes of broad-spectrum antibiotics. Clin Microbiol Infect 1997<br />

Feb;3 Suppl 4:S20-S31.<br />

41. Viaene E, Chanteux J, Servais H, et al. Comparative stability studies of antipseudomonal betalactams<br />

for potential administration through portable elastomeric pumps (home therapy for cystic<br />

fibrosis patients) and motor-operated syringes (intensive care units). Antimicrob Agents<br />

Chemother 2002 Aug;46(8):2327-32.<br />

42. Malik I, Shaharyar. Comparison of meropenem with ceftazidime as monotherapy of cancer<br />

patients with chemotherapy induced febrile neutropenia. J Pak Med Assoc 2002 Jan;52(1):15-8.<br />

43. Hou F, Li J, Wu G, et al. A randomized, controlled clinical trial of meropenem versus<br />

imipenem/cilastatin for the treatment of bacterial infections. Chin Med J (Engl) 2002<br />

Dec;115(12):1849-54.<br />

44. Manes G, Rabitti PG, Menchise A, et al. Prophylaxis with meropenem of septic complications in<br />

acute pancreatitis: a randomized, controlled trial versus imipenem. Pancreas 2003<br />

Nov;27(4):e79-83.<br />

45. Erdem I, Kucukercan M, Ceran N, et al. In vitro activity of combination therapy with cefepime,<br />

piperacillin/tazobactam, or meropenem with ciprofloxacin against multidrug resistant<br />

Pseudomonas aeruginosa strains. Chemotherapy 2003 Dec;49(6):294-7.<br />

159


46. Hung KC, Chiu HH, Tseng YC, et al. Monotherapy with meropenem versus combination therapy<br />

with ceftazidime plus amikacin as empirical therapy for neutropenia in children with malignancy.<br />

J Microbiol Immunol Infect 2003 Dec;36(4):254-9.<br />

47. Rhomberg PR, Jones RN, Sader HS, et al. Results from the meropenem yearly susceptibility test<br />

information (MYSTIC) Program: report of the 2001 data from United States medical centers. Int<br />

J Antimicrob Agents 2004 Jan;23(1):52-9.<br />

48. Verwaest C, Belgian Multicenter Study Group. Meropenem versus imipenem/cilastatin as<br />

empirical monotherapy for serious bacterial infections in the intensive care unit. Clin Microbiol<br />

Infect 2000 Jun;6(6):294-302.<br />

49. Rhomberg PR, Jones RN, Sader HS, et al. Antimicrobial resistance rates and clonality results<br />

from the meropenem yearly susceptibility test information collection (MYSTIC) Program: Report<br />

of the year five (2003). Diagn Microbiol Infect Dis 2004 Aug;49(4):273-81.<br />

50. Ciofu O, Jensen T, Pressler T, et al. Meropenem in cystic fibrosis patients infected with resistant<br />

Pseudomonas aeruginosa or Burkholderia cepacia and with hypersensitivity to beta-lactam<br />

antibiotics. Clin Microbiol infect 1996;2(2):91-98.<br />

51. Gilbert DN, Moellering RC, Eliopoulos GM, Sande MA. The Sanford guide to antimicrobial<br />

therapy. Thirty-Fourth Edition, 2004. Antimicrobial Therapy, Inc. Hyde Park, VT.<br />

52. Romanelli G, Cravarezza P, Pozzi A, et al. Carbapenems in the treatment of severe communityacquired<br />

pneumonia in hospitalized elderly patients: a comparative study against standard<br />

therapy. J Chemother 2002 Dec;14(6):609-17<br />

53. Nelson WK, Rayback PA, Quinones R, et al. Empiric carbapenem monotherapy in pediatric bone<br />

marrow transplant recipients. Ann Pharmacother 2002 Sep;36(9):1360-5.<br />

54. Hou F, Wu G, Zheng B. A randomized, controlled clinical trial of meropenem and<br />

imipenem/cilastatin in the treatment of acute bacterial infections. Zhonghua Nei Ke Za Zhi 2001<br />

Sep;40(9):589-93.<br />

55. Bartoloni A, Strohmeyer M, Corti G, et al. Multicenter randomized trial comparing meropenem<br />

(1.5g daily) and imipenem/cilastatin (2g daily) in the hospital treatment of community-acquired<br />

pneumonia. Drugs Exp Clin Res 1999;25(6):243-52.<br />

160


<strong>Pharmacotherapy</strong> <strong>Review</strong> of Chloramphenicol<br />

AHFS 081208<br />

October 27, 2004<br />

I. Overview<br />

In 1947, chloramphenicol was isolated from Streptomyces venezuelae, and was approved by the<br />

FDA in 1950. Chloramphenicol is now available synthetically for therapeutic use. 1 It is marketed<br />

as an injection, but the drug is seldom used today due to its potential toxicity and the availability<br />

of other antibiotics.<br />

Chloramphenicol is a true broad-spectrum antibiotic. It is active against a wide range of grampositive<br />

and gram-negative bacteria, many anaerobic bacteria, Chlamydia, and Rickettsia. It is<br />

inactive against fungi. In vitro concentrations of 0.1—20 mcg/mL of chloramphenicol are<br />

generally effective against susceptible strains. 1 Since hematologic toxicity can be dose-related,<br />

peak serum concentrations above 25 mcg/ml are discouraged.<br />

Injectable chloramphenicol is available as a generic formulation. This review encompasses all<br />

dosage forms and strengths. Table 1 lists the drugs included in this review.<br />

Table 1. Antibiotics in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name<br />

Chloramphenicol Sodium Succinate Injection Chloromycetin*<br />

*Generic Available.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Meningococcal Disease<br />

Chloramphenicol is an option for the treatment of meningococcal disease, including meningitis,<br />

according to the World Health Organization (WHO). 2 Chloramphenicol is a good choice, due to<br />

the drug’s ability to penetrate sufficiently into the cerebrospinal space. The WHO guidelines<br />

recommend either penicillin or ampicillin as the drug of choice, and ceftriaxone and cefotaxime as<br />

excellent alternatives. Although the intravenous route is recommended, clinical studies have<br />

shown the use of intramuscular chloramphenicol in oil to be as efficacious as intravenous<br />

ampicillin. Penetration of chloramphenicol into the cerebrospinal space has been reported to be<br />

good even after oral administration.<br />

Anthrax<br />

The antimicrobial susceptibility patterns of Bacillus anthracis isolates associated with the<br />

intentional exposures in 2001 were determined. 3 Isolates were susceptible to ciprofloxacin and<br />

doxycycline, the two drugs approved for post-exposure prophylaxis to B. anthracis. These strains<br />

were also susceptible to chloramphenicol, clindamycin, rifampin, vancomycin, and clarithromycin,<br />

but limited or no data exists regarding use of these agents in the treatment or prophylaxis of B.<br />

anthracis infections. To clarify, chloramphenicol is not formally FDA-approved for the treatment<br />

or prophylaxis of anthrax, it is an alternative therapy.<br />

As with all antibiotics, use of chloramphenicol is also dependent on the drug’s indications and<br />

spectrum of coverage.<br />

161


III.<br />

Indications of Chloramphenicol<br />

Chloramphenicol should not be used for the treatment of trivial infections, as a prophylactic agent<br />

to prevent bacterial infections, or when it is not indicated as in the treatment of colds, influenza, or<br />

throat infections. 4 Additionally, although not FDA-approved indications, chloramphenicol has<br />

been used in the following infections: Anthrax, Cholera, Melioidosis, Glanders, Burkholderia<br />

cepacia infections (Cystic fibrosis patients), Plague, Tularemia, Brucellosis, and Ehrlichiosis.<br />

Table 2. FDA-Approved Indications 4, 5<br />

Drug Serious Infections Acute Infections<br />

Chloramphenicol<br />

✔<br />

Those for which less potentially dangerous drugs are<br />

ineffective or contraindicated caused by susceptible strains of<br />

Salmonella species; H. influenzae, specifically, meningeal<br />

infections; rickettsiae; lymphogranuloma-psittacosis group;<br />

various gram-negative bacteria causing bacteremia, meningitis<br />

or other serious gram-negative infections; infections involving<br />

anaerobic organisms, when Bacteroides fragilis is suspected;<br />

other susceptible organisms which have been demonstrated to<br />

be resistant to all other appropriate antimicrobial agents.<br />

If presumptive therapy is initiated, perform in vitro sensitivity<br />

tests concurrently, so that the drug may be discontinued if less<br />

potentially dangerous agents are indicated.<br />

✔<br />

Those caused by S. typhi.<br />

Chloramphenicol is a drug of choice.<br />

In treatment of typhoid fever, some<br />

authorities recommend that<br />

chloramphenicol be used at<br />

therapeutic levels for 8 to 10 days<br />

after the patient becomes afebrile, to<br />

lessen the possibility of relapse. It is<br />

not recommended for the routine<br />

treatment of the typhoid “carrier<br />

state.”<br />

IV. Pharmacokinetic Parameters<br />

Chloramphenicol base is absorbed rapidly from the intestinal tract and is 75% to 90%<br />

bioavailable. In adults, at doses of 1g every 6 hours for 8 doses, the average peak serum level was<br />

11.2mcg/ml 1 hour after the first dose, and 18.4mcg/ml after the fifth 1g dose. Mean serum levels<br />

ranged from 8 to 14mcg/ml over the 48 hour period.<br />

Chloramphenicol diffuses rapidly, but its distribution is not uniform. Highest concentrations are<br />

found in liver and kidney, and lowest concentrations are found in brain and cerebrospinal fluid<br />

(CSF). However, chloramphenicol enters the CSF, even in the absence of meningeal<br />

inflammation, appearing in concentrations 45% to 99% of those found in the blood. Measurable<br />

levels are also detected in pleural and ascitic fluids, saliva, milk, and in the aqueous and vitreous<br />

humors.<br />

Table 3. Pharmacokinetic Parameters of Chloramphenicol 4, 5<br />

Drug Mechanism of<br />

Action<br />

Bioavailability<br />

Protein<br />

Binding<br />

Metabolism Active<br />

Metabolites<br />

Elimination<br />

Chloramphenicol Chloramphenicol 70-90% 60% Liver N o Total urinary<br />

binds to 50 S<br />

excretion<br />

ribosomal subunits of<br />

ranges from<br />

bacteria and interferes<br />

68-99% over<br />

with or inhibits protein<br />

3 days<br />

synthesis. In vitro,<br />

chloramphenicol<br />

exerts mainly a<br />

bacteriostatic effect on<br />

a wide range of gramnegative<br />

and grampositive<br />

bacteria.<br />

Half-<br />

Life<br />

4 hours


V. Drug Interactions<br />

Table 4 lists the most significant (Level 2) drug-drug interactions for chloramphenicol. No level 1<br />

interactions have been documented with chloramphenicol and other drugs.<br />

Table 4. Drug Interactions of Chloramphenicol 6<br />

Drug Significance Interaction Mechanism<br />

Chloramphenicol Level 2<br />

Delayed, moderate,<br />

Chloramphenicol and<br />

sulfonylureas<br />

suspected<br />

possible hypoglycemia.<br />

Level 2<br />

Delayed, moderate,<br />

suspected<br />

Level 2<br />

Delayed, moderate,<br />

suspected<br />

Level 2<br />

Delayed, moderate,<br />

suspected<br />

Chloramphenicol and<br />

anticoagulants<br />

Chloramphenicol and<br />

hydantoins<br />

Chloramphenicol and<br />

iron salts (ferrous<br />

fumerate, ferrous<br />

gluconate, ferrous<br />

sulfate, iron dextran, etc.)<br />

Reduction in sulfonylurea hepatic clearance by chloramphenicol<br />

leading to sulfonylurea accumulation (proposed mechanism), and<br />

Possible inhibition of hepatic metabolism of oral<br />

anticoagulants, causing the anticoagulant action of oral<br />

anticoagulants to be enhanced by chloramphenicol.<br />

Alteration in phenytoin metabolism can lead to increased serum<br />

phenytoin concentrations with potential toxicity.<br />

Chloramphenicol concentrations may also change.<br />

Decreased iron clearance and erythropoiesis due to direct bone<br />

marrow toxicity from chloramphenicol, resulting in increased<br />

serum iron levels.<br />

Additional less severe (Level 3 and Level 4) drug-drug interactions have been documented with<br />

chloramphenicol and include the following drugs 6 :<br />

• Vitamin B12 (Level 3)- The hematologic effects of vitamin B12 in patients with<br />

pernicious anemia may be decreased by concurrent administration of chloramphenicol.<br />

• Barbiturates (Level 4)- The metabolism of chloramphenicol may be increased and the<br />

barbiturate metabolism decreased. The efficacy of chloramphenicol may be reduced<br />

while the effects of the barbiturate are enhanced. The effects can persist for days after<br />

the barbiturate is withdrawn.<br />

• Penicillins (Level 4)- Mechanism is not fully understood. Synergistic effects may<br />

develop in the treatment of certain microorganisms, but antagonism has also been<br />

reported in animal studies.<br />

• Cyclophosphamide (Level 4)- Thought to result from inhibition of the hepatic<br />

microsomal enzyme system by chloramphenicol. The interaction may result in a<br />

decreased or delayed activation of cyclophosphamide. It is unclear at this time if a<br />

significant decrease in the effect of cyclophosphamide would result from this interaction.<br />

• Cyclosporine (Level 4)- Mechanism is unknown. Trough plasma concentrations of<br />

cyclosporine may be elevated, increasing the likelihood of adverse effects (e.g.<br />

nephrotoxicity).<br />

• Tacrolimus (Level 4)- Inhibition of hepatic metabolism of tacrolimus is suspected in this<br />

interaction. Tacrolimus concentrations may be elevated, increasing the risk of toxicity.<br />

• Rifampin (Level 4)- Chloramphenicol metabolism may be increased due to induction of<br />

hepatic microsomal enzymes by rifampin. Chloramphenicol serum levels progressively<br />

decrease, possibly leading to decreased anti-infective action. The effects could continue<br />

for days after rifampin is withdrawn.<br />

163


VI.<br />

Adverse Drug Events<br />

Chloramphenicol use is associated with a black box warning with the following important<br />

prescribing information.<br />

1, 4, 5<br />

Black Box Warning<br />

Serious and fatal blood dyscrasias (aplastic anemia, hypoplastic anemia, thrombocytopenia and<br />

granulocytopenia) occur after Chloramphenicol administration. Aplastic anemia, which later<br />

terminated in leukemia, has been reported. Blood dyscrasias have occurred after both short-term<br />

and prolonged therapy. Chloramphenicol must not be used when less potentially dangerous agents<br />

are effective. It must not be used to treat trivial infections (e.g., influenza, colds, throat<br />

infections), infections other than indicated, or as prophylaxis for bacterial infections.<br />

It is essential that adequate blood studies be performed during treatment. While blood studies may<br />

detect early peripheral blood changes, such as leukopenia, reticulocytopenia or granulocytopenia<br />

before they become irreversible, such studies cannot be relied upon to detect bone marrow<br />

depression prior to development of aplastic anemia. To facilitate appropriate studies and<br />

observation, patients should be hospitalized.<br />

Blood Dyscrasias<br />

Serious and fatal blood dyscrasias (aplastic anemia, hypoplastic anemia, thrombocytopenia and<br />

granulocytopenia) occur. 5 An irreversible type of marrow depression leading to aplastic anemia<br />

with a high rate of mortality is characterized by the appearance of bone marrow aplasia or<br />

hypoplasia weeks or months after therapy. Peripherally, pancytopenia is most often observed, but<br />

only one or two of the three major cell types (erythrocytes, leukocytes and platelets) may be<br />

depressed. This complication appears unrelated to administration route. One estimate, based on<br />

149 cases, stated that the route was oral in 83%, parenteral in 14% and rectal in 3%. Several cases<br />

of aplastic anemia have been associated even with chloramphenicol ophthalmic ointment.<br />

A dose-related reversible type of bone marrow depression may occur and is associated with<br />

sustained serum levels at peak ≥25mcg/ml, trough ≥10mcg/ml. This type of marrow depression is<br />

characterized by vacuolization of the erythroid cells, a decrease in red cell iron uptake, an increase<br />

in circulating serum iron with saturation of iron-binding globulin (usually within 6 to 10 days) and<br />

reduction of reticulocytes (usually within 5 to 7 days)and leukopenia; it responds promptly to<br />

withdrawal of the drug.<br />

Table 5 lists the common adverse events for chloramphenicol.<br />

164


Table 5. Common Adverse Events (%) Reported for Chloramphenicol 6<br />

Adverse Event<br />

Chloramphenicol<br />

Body as a Whole<br />

Malaise<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Glossitis/stomatitis<br />

Appetite decrease<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

Mild Depression<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Hematologic<br />

Neutropenia<br />

Agranulocytosis<br />

Bone marrow depression<br />

Aplastic anemia (later as leukemia)<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

Other<br />

Angioedema<br />

Convulsions<br />

Optic and peripheral neuritis<br />

Gray syndrome*<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

1:40,000 cases<br />

✔ Adverse event reported; specific percentages not available.<br />

*Toxic reactions including fatalities (approximately 40%) have occurred in premature infants and newborns; the signs and<br />

symptoms associated with these reactions have been referred to as the “gray syndrome”.<br />

✔<br />

✔<br />

✔<br />

165


VII.<br />

Dosing and Administration<br />

1, 4, 5<br />

Table 6. Dosing for Chloramphenicol<br />

Drug Availability Dose /Frequency/Duration<br />

Chloramphenicol<br />

Powder for Injection:<br />

1g (100mg/ml when<br />

reconstituted) vials<br />

Therapeutic concentrations should be maintained as follows: Peak 10 to 20mcg/ml;<br />

trough 5 to 10mcg/ml.<br />

Monitoring:<br />

It is important to monitor serum levels because of the variability of<br />

chloramphenicol's pharmacokinetics. Monitor serum concentrations weekly; monitor<br />

more often in patients with hepatic dysfunction, in therapy >2 weeks or with<br />

potentially interacting drugs.<br />

Adults:<br />

50mg/kg/day in divided doses every 6 hours for typhoid fever and rickettsial<br />

infections. Exceptional infections (e.g., meningitis, brain abscess) due to moderately<br />

resistant organisms may require dosage up to 100mg/kg/day to achieve blood levels<br />

inhibiting the pathogen; decrease high doses as soon as possible.<br />

Children:<br />

50 to 75mg/kg/day in divided doses every 6 hours are recommended for most<br />

indications. For meningitis, 50 to 100mg/kg/day in divided doses every 6 hours has<br />

been recommended.<br />

Newborns:<br />

(See Gray syndrome under Adverse Reactions.) 25mg/kg/day in 4 doses every 6<br />

hours usually produces and maintains adequate concentrations in blood and tissues.<br />

Give increased dosage demanded by severe infections only to maintain the blood<br />

concentration within an effective range. After the first 2 weeks of life, full-term<br />

infants ordinarily receive up to 50mg/kg/day in 4 doses every 6 hours.<br />

• Neonates (2kg):<br />

25mg/kg once daily.<br />

• Neonates over 7 days (>2kg):<br />

50mg/kg/day in divided doses every 12 hours.<br />

These dosage recommendations are extremely important because blood concentration<br />

in all premature and full-term infants


Special Dosing Considerations<br />

1, 5, 7<br />

Table 7. Special Dosing Considerations for Chloramphenicol<br />

Drug<br />

Hepatic Pediatric Use<br />

Chloramphenicol<br />

Renal<br />

Dosing<br />

Dosing<br />

Yes<br />

Impaired metabolic<br />

processes require that<br />

doses be adjusted based<br />

on drug concentration in<br />

the blood. An initial<br />

loading dose of 1g<br />

followed by 500mg every<br />

6 hours has been<br />

recommended in<br />

impaired hepatic<br />

function.<br />

Yes<br />

See adverse<br />

event section for<br />

warning on Gray<br />

syndrome<br />

Pregnancy<br />

Category<br />

C<br />

Can Drug Be Crushed/Stability<br />

Following reconstitution,<br />

chloramphenicol at 100mg/ml is<br />

stable for 30 days at room<br />

temperature.<br />

VIII.<br />

Comparative Effectiveness<br />

Since chloramphenicol is an older antibiotic, limited recent clinical comparable data is available.<br />

Table 8 describes clinical studies that have recently looked at the efficacy and place-in-therapy of<br />

chloramphenicol.<br />

Table 8. Additional Outcomes Evidence for Chloramphenicol<br />

Study Sample Treatment /<br />

Duration<br />

Third generation<br />

cephalosporins<br />

vs. conventional<br />

antibiotics for<br />

acute bacterial<br />

meningitis 8<br />

Meta-analysis<br />

of 18 trials<br />

and 993<br />

patients<br />

Criteria for trials<br />

included<br />

randomized,<br />

controlled studies<br />

comparing<br />

ceftriaxone or<br />

cefotaxime with<br />

conventional<br />

antibiotics<br />

Treatment of<br />

Rickettsia with<br />

five different<br />

antibiotic<br />

regimens 9 n=87 Doxycycline,<br />

chloramphenicol,<br />

ciprofloxacin,<br />

doxycycline plus<br />

chloramphenicol,<br />

and doxycycline<br />

167<br />

Results<br />

The goal of this study was to determine the effectiveness and safety of the third<br />

generation cephalosporins versus conventional treatment with<br />

penicillin/ampicillin-chloramphenicol in patients with community-acquired<br />

acute bacterial meningitis:<br />

• There was no heterogeneity of results among the studies in any<br />

outcome except diarrhea.<br />

• There was no statistically significant difference between the groups<br />

in the risk of death (risk difference -1%; 95% confidence interval<br />

(CI) -4% to +3%), risk of deafness (risk difference -4%; 95% CI -<br />

9% to +1%), or risk of treatment failure (risk difference -2%; 95%<br />

CI -5% to +2%).<br />

• There was a significant decrease in the risk of culture positivity of<br />

CSF after 10-48 hours (risk difference -6%; 95% CI -11% to 0%)<br />

and statistically significant increase in the risk of diarrhea between<br />

the groups (risk difference +8%; 95% CI +3% to +13%) with the<br />

third generation cephalosporins.<br />

• The risks of neutropenia and skin rash were not significantly<br />

different between the two groups.<br />

• All the studies were conducted in the 1980’s except two, which<br />

were conducted in 1993 and 1996.<br />

• Summary: Although the review shows no clinically important<br />

difference between ceftriaxone or cefotaxime and conventional<br />

antibiotics, the studies were done decades ago and may not apply<br />

to current routine practice. However, in situations where<br />

ceftriaxone or cefotaxime are not available or affordable,<br />

ampicillin-chloramphenicol combination may be used as an<br />

alternative.<br />

In examining the clinical effectiveness of five different antibiotic regimens for<br />

infection with Rickettsia, in terms of duration of fever:<br />

• The mean time to defervescence was 2.9 days for doxycycline, 4.0<br />

days for chloramphenicol, and 4.2 days for ciprofloxacin.<br />

• In patients receiving combinations of doxycycline plus<br />

chloramphenicol and doxycycline plus ciprofloxacin, fever subsided in


Resistance to<br />

chloramphenicol<br />

among<br />

vancomycin<br />

resistant<br />

enterococcal<br />

(VRE)<br />

infections 10<br />

All VRE<br />

isolates were<br />

analyzed for<br />

susceptibility<br />

profiles<br />

plus ciprofloxacin<br />

10-year study<br />

period (1991-<br />

2000)<br />

3.4 and 4.0 days, respectively.<br />

• The outcome was favorable in all patients, and no deaths or relapses<br />

were observed within two months.<br />

In evaluating the antimicrobial susceptibility profiles of all VRE blood isolates<br />

from 1991-2000, and to look at hospital use of specific antibiotics and antibiotic<br />

classes and chloramphenicol resistant vancomycin-resistant enterococcal (CR-<br />

VRE) prevalence:<br />

• Chloramphenicol is effective when VRE isolates are susceptible to this<br />

agent.<br />

• During the 10-year study period, the prevalence of CR-VRE increased<br />

from 0 to 11% ( p< 0.001, trend).<br />

• CR-VRE prevalence was correlated only with chloramphenicol use<br />

(p=0.05) and quinolone use (p= 0.01).<br />

• If these trends continue, dependence on newer, more expensive agents<br />

will increase. The correlation between both chloramphenicol use and<br />

quinolone use and the prevalence of CR-VRE suggests that efforts to<br />

preserve the utility of chloramphenicol in VRE infections may depend<br />

-<br />

on optimizing the use of these agents.<br />

In vitro postantibiotic<br />

effect<br />

Two stains of<br />

Bacillus<br />

- In investigating the in vitro post-antibiotic effect (PAE) of 19 antibacterial<br />

agents against two strains of Bacillus anthracis:<br />

of 19 drugs/drug<br />

classes on<br />

Bacillus<br />

anthracis 11<br />

anthracis<br />

were used<br />

• The PAE was determined by calculating the time required for the<br />

viable counts of antibiotic-exposed bacteria (at concentrations of 10x<br />

MIC and exposure for 2h) at 37 degrees C to increase by 1 log10<br />

above the counts observed immediately after antibiotic removal<br />

compared with the corresponding time for controls not exposed to<br />

antibiotics.<br />

• The PAEs of the fluoroquinolones (ciprofloxacin, ofloxacin,<br />

levofloxacin, moxifloxacin and garenoxacin*) were 2-5h.<br />

• The macrolide (erythromycin, clarithromycin and telithromycin) PAEs<br />

were 1-4h and that of clindamycin was 2h.<br />

• The PAEs induced by tetracycline and minocycline were 1-3h.<br />

• The PAEs induced by the beta-lactams (penicillin G, amoxicillin and<br />

ceftriaxone), vancomycin, linezolid, and chloramphenicol were 1-2h.<br />

The PAE induced by rifampicin was 4-5h.<br />

• Quinupristin/dalfopristin had the longest PAE, lasting for 7-8h.<br />

• These results indicate that the PAE is unrelated to the MIC but may be<br />

related to the rapidity of bacterial kill. These observations may bear<br />

importance on treatment regimens of human anthrax.<br />

Chloramphenicol n=101 In evaluating the trends in prevalence of chloramphenicol resistance in VRE<br />

quinolone use 12<br />

resistance in VRE<br />

bacteremia:<br />

blood isolates from 1991-2002 to identify risk factors for chloramphenicol<br />

resistance among isolates:<br />

impact of prior<br />

• During the review period, the annual prevalence of<br />

Chloramphenicol<br />

vs. rifampin for<br />

Streptococcus<br />

pneumoniae-<br />

A group of<br />

107 isolates<br />

of S.<br />

pneumoniae<br />

The min.<br />

inhibitory<br />

concentration<br />

(MIC) and min.<br />

chloramphenicol-resistant VRE increased from 0% to 12% (p <<br />

.001, chi-square test for trend).<br />

• Independent risk factors for chloramphenicol-resistant VRE were<br />

prior chloramphenicol use (odds ratio [OR], 10.9; 95% confidence<br />

interval [CI95], 1.72-68.91; P = .01) and prior fluoroquinolone use<br />

(OR, 4.74; CI95, 1.15-19.42; p = 0.03).<br />

• Chloramphenicol-resistant VRE isolates were more likely to be<br />

susceptible to beta-lactams and resistant to tetracycline than were<br />

chloramphenicol-susceptible VRE isolates.<br />

• Significant increases in the prevalence of chloramphenicolresistant<br />

VRE may limit the future utility of chloramphenicol in<br />

the treatment of VRE infections, and close monitoring of<br />

susceptibility trends should continue. The association between<br />

fluoroquinolone use and chloramphenicol-resistant VRE,<br />

reflecting possible co-selection of resistance, suggests that recent<br />

dramatic increases in fluoroquinolone use may have broader<br />

implications than previously recognized.<br />

In evaluating the minimal inhibitory concentration and minimal bactericidal<br />

concentration for chloramphenicol and rifampin in strains of S. pneumoniae:<br />

• Chloramphenicol resistance was found in 21% of the 107 isolates.<br />

• In the group susceptible to penicillin, 11% were resistant to<br />

168


induced<br />

meningitis and<br />

systemic<br />

infections 13<br />

from children<br />

under 5<br />

*Not available in the United States.<br />

bacterial<br />

concentration<br />

(MBC) for<br />

chloramphenicol<br />

and rifampin were<br />

obtained<br />

chloramphenicol, and in the group resistant to penicillin, 28% were<br />

resistant to chloramphenicol as well.<br />

• MBC was found >4microg/ml in 28% of the isolates susceptible to<br />

penicillin and in 60% of the resistant isolates.<br />

• No isolates were found resistant to rifampin.<br />

• However, 2 penicillin resistant isolates showed CBM >1microg/ml to<br />

rifampin, and one with CIM = 1microg/ml had a MBC to rifampicin of<br />

16microg/ml.<br />

• Meningitis isolates showed higher CIM and CBM than the group of<br />

total isolates.<br />

• These data suggest that chloramphenicol is not recommended for<br />

invasive infections caused by S. pneumoniae in Colombia. Rifampin<br />

is a more effective therapy in combination with other antibiotics for<br />

treatment of this kind of infections. Further studies are necessary to<br />

clarify the significance of low levels of MBC to rifampin found in<br />

some strains, since this may affect the efficacy of therapies that<br />

include this antibiotic.<br />

Additional Evidence<br />

Dose Simplification: Since chloramphenicol is only available in a single formulation, there isn’t<br />

another agent or dosage formulation for which chloramphenicol can be compared to for dose<br />

simplification. Therefore, dose simplification does not apply to this particular class.<br />

Stable Therapy: Not applicable.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal clinical data<br />

pertinent to physician visits or hospitalizations with chloramphenicol.<br />

IX.<br />

Conclusions<br />

Use of chloramphenicol should be reserved for serious infections or infections resistant to other<br />

therapies. Use is likely limited to hospitalized patients, where intravenous therapy can be given<br />

and monitored appropriately. Because of the black box warning associated with chloramphenicol<br />

use, and due to the fact that the drug should not be used for trivial infections, there is not a role for<br />

the use of chloramphenicol in general use. Chloramphenicol should be available for special<br />

needs/circumstances that require medical justification through the prior authorization process.<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the<br />

generics in the class and offer no significant clinical advantage over other alternatives in general<br />

use.<br />

X. Recommendations<br />

No brand of chloramphenicol is recommended for preferred status.<br />

169


References<br />

1. Clinical Pharmacology, 2004. Available on-line at: www.cp.gsm.com. Accessed September 15,<br />

2004.<br />

2. World Health Organization. Control of epidemic meningococcal disease. WHO practical<br />

guidelines. 2 nd edition. Available at: www.who.int/emc. Accessed September 21, 2004.<br />

3. Centers for Disease Control. Health Advisory. Antimicrobial susceptibility of Bacillus anthracis<br />

isolates associated with intentional distribution in Florida, New Jersey, New York, Pennsylvania,<br />

Virginia, and Washington, D.C., September-October, 2001. Available at: www.cdc.gov.<br />

Accessed September 21, 2004.<br />

4. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

5. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

6. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

7. Briggs GG, Freeman RK, Yaffe SJ. Drugs in pregnancy and lactation. A reference guide to fetal<br />

and neonatal risk. Sixth Edition. Lippincott, Williams, & Wilkins, Philadelphia, 2002.<br />

8. Prasad K, Singhal T, Jain N, et al. Third generation cephalosporins versus conventional<br />

antibiotics in treating acute bacterial meningitis. Cochrane Database Syst Rev<br />

2004;(2):CD001832.<br />

9. Gikas A, Doukakis S, Pediaditis J, et al. comparison of the effectiveness of five different<br />

antibiotic regimens on infection with Rickettsia typhi: therapeutic data from 87 cases. Am J Trop<br />

Med Hyg 2004 May;70(5):576-9.<br />

10. Lautenbach E, Gould CV, LaRosa LA, et al. Emergence of resistance to chloramphenicol among<br />

vancomycin resistant enterococcal (VRE) bloodstream isolates. Int J Antimicrob Agents 2004<br />

Feb;23(2):200-3.<br />

11. Athamna A, Athmana M, Medlej B, et al. In vitro post-antibiotic effect of fluoroquinolones,<br />

macrolides, beta-lactams, tetracyclines, vancomycin, clindamycin, linezolid, chloramphenicol,<br />

quinupristin/dalfopristin and rifampin on Bacillus anthracis. J Antimicrob Chemother 2004<br />

Apr;53(4):609-15.<br />

12. Gould CV, Fishman NO, Nachamkin I, et al. Chloramphenicol resistance in vancomycin –<br />

resistant enterococcal bacteremia: impact of prior fluoroquinolone use. Infect Control Hosp<br />

Epidemiol 2004 Feb;25(2):138-45.<br />

13. Hernandez M, Mejia GI, Trujillo H, et al. Effectiveness of the antibiotics chloramphenicol and<br />

rifampin in the treatment of Streptococcus pneumoniae-induced meningitis and systemic<br />

infections. Biomedica 2003 Dec;23(4):456-61.<br />

170


I. Overview<br />

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Single Entity Penicillins<br />

AHFS 081216<br />

October 27, 2004<br />

Penicillins are natural or semisynthetic antibiotics produced or derived from Penicilium fungus. 1<br />

The drugs are ß-lactam antibiotics structurally and chemically related to cephalosporins and<br />

cephamycins. Currently available penicillins can be divided into four groups primarily based on<br />

their spectrum of activity:<br />

o Natural Penicillins<br />

o Penicillinase-Resistant Penicillins<br />

o Aminopenicillines<br />

o Extended-Spectrum Penicillins<br />

NATURAL PENICILLINS<br />

• Penicillin G<br />

• Penicillin V<br />

Natural penicillins are produced by fermentation of mutant strains of Penicillium chrysogenum.<br />

Several natural penicillins have been produced, however, only penicillin G and Penicillin V are<br />

currently available. Natural penicillins are active in vitro against many gram-positive cocci<br />

including nonpenicillinase-producing Staphylococcus aureus and S. epidermidis, Streptococcus<br />

pneumoniae, groups A, B, C, G, H, K and L streptococci, viridians streptococci and certain strains<br />

of enterococci. Natural penicillins are hydrolyzed by staphylococcal penicillinases and are<br />

inactive against penicillinase-producing strains of S. aureus and S. epidermidis.<br />

Natural penicillins are also active against gram-negative cocci and some gram-negative bacilli.<br />

These pathogens include; Neisseria meningitides and most strains of nonpenicillinase-producing<br />

N. gonorrhoeae. The drugs are active in vitro against some gram-negative stains of Haemophilus<br />

influenzae, Pasteurella multocida, and Spiillum minus. The drugs are also active against most<br />

spirochets, including Treponema pallidum, T. pertenue, leptospira, Borrelia recurrentis, and B.<br />

burgdorferi, the causative pathogen of Lyme disease. 1<br />

PENICILLINASE-RESISTANT PENICILLINS<br />

• Dicloxacillin<br />

• Nafcillin<br />

• Oxacillin<br />

Penicillinase-resistant penicillins are stable against hydrolysis of most staphylococcal<br />

penicillinases, including S. aureus and S. epidermidis that are resistant to natural penicillins,<br />

aminopenicillins, and extended-spectrum penicillins. They have some activity against gramnegative<br />

bacteria and spirochetes, although they are less active against these organisms than<br />

natural penicillins. 1<br />

171


AMINOPENICILLINS<br />

• Amoxicillin<br />

• Ampicillin<br />

Aminopenicillins have enhanced activity against gram-negative bacteria compared with natural<br />

penicillins and penicillinase-resistant penicillins. Aminopenicillins are generally active against<br />

gram-positive aerobic bacilli. Similar to natural penicillins and extended-spectrum penicillins,<br />

aminopenicillins are readily hydrolyzed by staphylococcal penicillinases and are inactive against<br />

penicillinase-producing strains of S. aureus and S. epidermidis.<br />

Aminopenicillins are generally active in vitro against gram-negative cocci and bacilli as well as<br />

anaerobic bacteria. Also, aminopenicillins are active versus Enterobacteriaceae including some<br />

strains of E. coli, Proteus mirabilis, Salmonella, and Shigella.<br />

Clavulanic acid and sulbactam can inhibit certain ß-lactamases that generally inactivate<br />

aminopenicillins. Combination of amoxicillin and clavulanate potassium and combinations of<br />

ampicillin and sulbactam sodium are active against many ß-lactamase-producing organisms that<br />

are resistant of aminopenicillins alone. 1<br />

EXTENDED-SPECTRUM PENICILLINS<br />

• Piperacillin<br />

• Ticarcillin<br />

• Carbenicillin<br />

• Mezlocillin<br />

Extended-Spectrum Penicillins have a wider spectra that the aforementioned agents. They are<br />

more active against gram-negative aerobic and gram-negative anaerobic bacilli than<br />

aminopenicillins. The use of extended-spectrum penicillins are limited to treatment of serious<br />

infections caused by gram-negative bacilli and mixed aerobic-anaerobic bacterial infections.<br />

In vitro, they are generally active gram-positive and gram-negative cocci. Like natural penicillins<br />

and aminopenicillins, extended-spectrum penicillins are hydrolyzed by penicillinases and are<br />

therefore inactive against penicillinase-producing strains of S. aureus and S. epidermidis. The<br />

drugs are also active against strains of Enterobacteriaceae and certain strains of Pseudomonas.<br />

This review encompasses all dosage forms and strengths of the single entity penicillins. Table 1<br />

lists the drugs included in this review.<br />

172


Table 1. Single Entity Penicillins in this <strong>Review</strong> 1,2<br />

Generic Name Formulation Example Brand Name (s)<br />

Natural Penicillins<br />

Penicillin G (sodium and Sterile Suspension, Injection Bicillin LA, Bicillin CR *Pfizerpen,<br />

procaine)<br />

Wycillin<br />

Penicillin V<br />

Solution, Tables, Film-Coated *Penicillin VK, *Ledercillin VK,<br />

Tablets<br />

*Veetids<br />

Penicillinase-Resistant Penicillins<br />

Dicloxacillin Suspension, Capsules *Dynapen<br />

Nafcillin Injection *Nallpen, *Unipen<br />

Oxacillin Solution, Capsules, Injection *Bactocill<br />

Aminopenicillins<br />

Amoxicillin<br />

Chewable Tablets, Capsules, *Trimox, *Amoxil, Dispermox<br />

Suspension, Film-Coated Tablets<br />

Ampicillin Capsules, Injection, Suspension *Principen, Totacillin-N<br />

Extended-Spectrum Penicillins<br />

Carbenicillin Film-Coated Tablets Geocillin<br />

Piperacillin Injection *Pipracil<br />

Ticarcillin Injection Ticar<br />

*Generic Available.<br />

II. Evidence Based Medicine and Current Treatment Guidelines 1,3<br />

Penicillins continue to be the drugs of choice or alternatives for a variety of microorganisms.<br />

Methicillin is the drug of choice for sensitive strains of Staphylococcal infections. The<br />

aminopencillins are also used first-line for Enterococcus species, particular urinary tract infections<br />

caused by Enterococcus faecalis. Sensitive strains for Streptococcal infection including,<br />

pneumoniae, viridans group and groups A, B, C, G are generally treated empirically with<br />

Penicillin G or ampicillin. Clostridium perfringens is also treated with Penicillin G in<br />

combination with clindamycin. Anthrax (Bacillus anthracis) can also be alternatively treated with<br />

doses of Penicillin G or Amoxicillin. 4<br />

Gram-negative microorganisms such as Escherichia coli, Acinetobacter spp. and Pseudomonas<br />

aeruginosa are initially treated with combination penicillins such as amoxicillin/clavulanate,<br />

ampicillin/sulbactam or piperacillin/tazobactam. In the case of Pseudomonas aeruginosa,<br />

penicillins are usually combined with other antibiotics. Drugs of choice for Pasteurella multocida<br />

and Proteus mirabilis are Penicillin G and Ampicillin, respectively. Treponema pallidum, the<br />

causative microorganism responsible for Neurosyphilis, is treated with Penicillin G.<br />

Pneumonia<br />

Community acquired pneumonia can be caused by various microorganisms.<br />

Amoxicillin/clavulanate is considered an alternative option for the treatment on non-hospitalized<br />

community acquired pneumonia. Patients hospitalized with severe community-acquired<br />

pneumonia are treated empirically with ß-lactam inhibitors in combination with a fluoroquinolone<br />

or a macrolide. Nosocomial pneumonia is usually a mixed infection with anaerobic activity.<br />

Combination agents such as ticarcillin/clavulanate and piperacillin/tazobactam are generally given<br />

in combination with other agents, particularly when pseudomonas is suspected.<br />

Urinary Tract Infection<br />

ß-lactams such as amoxicillin or ampicillin are usually considered alternatives for uncomplicated<br />

cystitis. These agents are categorized as a Pregnancy Category B and can be therefore used in<br />

pregnancy. Extended-spectrum penicillins are administered for 14-day treatment of complicated<br />

urinary tract infections.<br />

173


Skin & Soft Tissue Infections<br />

Acute Cellulitis and Erysipelas caused by S. pyogens or S. aureus are generally treated with antistaphylococcal<br />

penicillins (nafcillin, oxacillin, or dicloxacillin). Necrotizing fasciitis due to mixed<br />

microorganisms, including anaerobes is empirically treated with ß-lactam inhibitor combinations.<br />

Diabetic foot infections are generally polymicrobial involving two to six different<br />

microorganisms. This infection requires broad coverage with ß-lactam inhibitors combination<br />

agents like piperacillin/tazobactam.<br />

Endocarditis Prophylaxis<br />

Endocarditis is a result of bacterial vegetation in the heart valves. Usual suspects include grampositive<br />

organisms such as Streptococci, S. aureus, Enterococci and coagulase-negative<br />

staphylococci. Amoxicillin is the drug of choice for prophylaxis. For patients unable to take oral<br />

medications, ampicillin is an alternative in either IM or IV formulation.<br />

Bacterial Meningitis<br />

Several penicillins are listed as drugs of choice for several pathogens responsible for bacterial<br />

meningitis. Penicillin G in combination with rifampin is indicated for meningitis caused by N.<br />

meningitides. Ampicillin and oxacillin are indicated for L. monocytogenes and methicillinsensitive<br />

S. aureus, respectively.<br />

Otitis Media<br />

Initial cases of otitis media in children can be treated with standard doses of amoxicillin. High<br />

doses of amoxicillin (80-100mg/kg/day) are used for treatment failures or recurrent episodes.<br />

Amoxicillin/clavulanate is also a treatment regimen for cases of otitis media.<br />

174


III.<br />

Susceptibility of the Single Entity Penicillins<br />

Table 2. Organisms Generally Susceptible to Single Entity Penicillins In Vitro 2<br />

Organisms Natural Penicillins Penicillinase-Resistant Penicillins Aminopenicillins Extended-Spectrum Penicillins<br />

Gram-<br />

Penicillin Penicillin Cloxacillin∞ Dicloxacillin Nafcillin Oxacillin Amoxicillin Ampicillin Carbenicillin Mezlocillin† Piperacillin Ticarcillin<br />

✔ = Generally<br />

Positive<br />

G V<br />

Susceptible<br />

Staphylococci<br />

✔* ✔* ✔ ✔ ✔ ✔ ✔* ✔* ✔* ✔* ✔*<br />

Staphylococcus<br />

aureus<br />

Staphylococcus<br />

Epidermidis<br />

Streptococci<br />

Streptococcus<br />

Pneumoniae<br />

Beta-hemolytic<br />

streptococci<br />

Enterococcus<br />

faecalis<br />

Streptococcus<br />

viridans<br />

Corynebaacterium<br />

diphtheriae<br />

Bacillus anthracis<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔* ✔* ✔* ✔*<br />

✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔<br />

✔ ✔<br />

Streptococcus<br />

agalactiae<br />

Streptococcus<br />

pyogens<br />

Erysipelothrix<br />

✔<br />

rhusiopathiae<br />

Listeria<br />

✔ ✔<br />

monocytogenes<br />

* Non-penicillinase producing<br />

† Mezlocillin has been discontinued in the United States and is not being reviewed with this class.<br />

∞ Cloxacillin is no longer available in the United States and is not listed in the Alabama Medicaid drug file. It is not included in this review.<br />

175


Table 2. Continued…Organisms Generally Susceptible to Single Entity Penicillins In Vitro 2<br />

Organisms Natural Penicillins Penicillinase-Resistant Penicillins Aminopenicillins Extended-Spectrum Penicillins<br />

Gram-<br />

Penicillin Penicillin Cloxacillin∞ Dicloxacillin Nafcillin Oxacillin Amoxicillin Ampicillin Carbenicillin Mezlocillin† Piperacillin Ticarcillin<br />

✔ = Generally<br />

Negative<br />

G V<br />

Susceptible<br />

Escherichia coli<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

Haemophilus<br />

influenzae<br />

✔ ✔ ✔ ✔ ✔ ✔<br />

Haemophilus<br />

parainfluenzae<br />

✔<br />

Eikenella<br />

corrodens<br />

Bacteroides<br />

melaninogenicus<br />

Klebsiella sp.<br />

✔ ✔ ✔<br />

Neisseria<br />

gonorrhoeae<br />

Neisseria<br />

meningitides<br />

Proteus mirabilis<br />

Salmonella sp.<br />

Shigella sp.<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔<br />

Morganella<br />

morganii<br />

✔ ✔ ✔ ✔<br />

Proteus vulgaris<br />

✔ ✔ ✔ ✔<br />

Providencia<br />

stuartii<br />

Enterobacter sp.<br />

Citrobacter sp.<br />

✔ ✔ ✔ ✔ ✔<br />

✔<br />

Pseudomonas<br />

aeruginosa<br />

✔ ✔ ✔ ✔<br />

Serratia sp.<br />

✔ ✔ ✔ ✔<br />

Acinetobacter sp.<br />

✔ ✔ ✔<br />

Streptobacillus<br />

moniliformis<br />

✔ ✔<br />

∞ Cloxacillin is no longer available in the United States and is not listed in the Alabama Medicaid drug file. It is not included in this review.<br />

176


Table 2. Continued…Organisms Generally Susceptible to Single Entity Penicillins In Vitro 2<br />

Anaerobic<br />

Penicillin Penicillin Cloxacillin∞ Dicloxacillin Nafcillin Oxacillin Amoxicillin Ampicillin Carbenicillin Mezlocillin† Piperacillin Ticarcillin<br />

✔ = Generally<br />

G V<br />

Susceptible<br />

Clostridium sp.<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

Peptococcus sp.<br />

Peptostreptococcus<br />

sp.<br />

Bacteroides sp.<br />

Fusobacterium sp.<br />

Eubacterium sp.<br />

Treponema<br />

pallidum<br />

Actinomyces bovis<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔<br />

✔ ✔<br />

✔ ✔ ✔<br />

Veillonella sp.<br />

✔ ✔ ✔<br />

† Mezlocillin has been discontinued in the United States and is not being reviewed with this class.<br />

∞ Cloxacillin is no longer available in the United States and is not listed in the Alabama Medicaid drug file. It is not included in this review.<br />

177


III.<br />

Indications of the Single Entity Penicillins<br />

Table 3. FDA Approved Indications of the Single Entity Penicillins<br />

Drug Meningococ<br />

cal<br />

Meningitis<br />

Septicemia Actinomycosis Clostridial<br />

Infections<br />

Fusospirochetal<br />

Infections<br />

Rat<br />

Bite<br />

Fever<br />

Listeria<br />

Infections<br />

Penicillin G X X X X X X X Endocarditis, Pasteurella<br />

inf., Erysipeloid,<br />

Diphtheria, Anthrax,<br />

streptococcal inf.,<br />

empyema, pneumonia,<br />

pericarditis, syphilis,<br />

neurosyphilis, disseminated<br />

gonococcal inf.<br />

Penicillin V X Staphylococcal,<br />

streptococcal, and<br />

pneumococcal inf., to<br />

prevent recurrence<br />

following rheumatic fever<br />

or chorea.<br />

Dicloxacillin<br />

Nafcillin<br />

Oxacillin<br />

Amoxicillin<br />

Ampicillin<br />

X<br />

Injection only<br />

X<br />

Injection<br />

only<br />

Other<br />

Treatment of infections<br />

caused by penicillinaseproducing<br />

staphylococci.<br />

Treatment of infections<br />

caused by penicillinaseproducing<br />

staphylococci<br />

that have demonstrated<br />

susceptibility to the drug.<br />

Treatment of infections<br />

caused by penicillinaseproducing<br />

staphylococci.<br />

Ear, nose and throat inf.,<br />

GU tract, skin and skin<br />

structure inf., lower RTI,<br />

and acute uncomplicated<br />

gonorrhea.<br />

Endocarditis, RTI inf., GI<br />

and GI inf.<br />

Carbenicillin<br />

UTI, prostatitis, and other<br />

infections caused by<br />

susceptible inf.<br />

Piperacillin X Mixed infections caused by<br />

susceptible inf., intraabdominal<br />

inf., UTI, GYN<br />

inf., lower RTI, skin and<br />

skin structure inf., bone and<br />

joint inf., gonococcal inf.,<br />

streptococcal inf.,<br />

prophylaxis for surgical<br />

procedures.<br />

Ticarcillin X Skin and skin structure inf.,<br />

acute and chromic RTI<br />

including P. aeruginosa,<br />

Proteus sp, and E. coli.<br />

Also GU inf., and inf.<br />

Caused by susceptible<br />

anaerobic bacteria.


IV.<br />

Pharmacokinetic Parameters of the Single Entity Penicillins<br />

Table 3 lists the pharmacokinetic parameters of the single entity penicillin antibiotics. Penicillins<br />

inhibit the biosynthesis of cell wall mucopeptide.<br />

Table 4. Pharmacokinetic Parameters of the Single Entity Penicillins 2, 5<br />

Drug Acid Stable Penicillanaseresistant<br />

% Protein<br />

Binding<br />

Elimination May be taken<br />

with meals<br />

NATURAL PENICILLINS<br />

Penicillin G No No 60 Renal IM/IV use<br />

Penicillin V No No 80 Renal Yes<br />

PENICILLINASE-RESISTANT<br />

Dicloxacillin Yes Yes 98 Renal No<br />

Nafcillin Yes Yes 87-90 Hepatic No<br />

Oxacillin Yes Yes 94 Hepatic No<br />

AMINOPENICLLINS<br />

Amoxicillin Yes No 20 Renal Yes<br />

Ampicillin Yes No 20 Renal No<br />

EXTENDED-SPECTRUM<br />

Carbenicillin Yes No 50 Renal No<br />

Piperacillin IM/IV use No 16 Renal IM/IV use<br />

Ticarcillin IM/IV use No 45 Renal IM/IV use<br />

V. Drug Interactions of the Single Entity Penicillins<br />

Table 5. Drug Interactions of the Single Entity Penicillins 6<br />

Drug Significance Interaction Mechanism<br />

Penicillins<br />

Level 1 (delayed, major,<br />

Tetracyclines impair the bacteriocidal effects of<br />

Tetracyclines<br />

suspected)<br />

penicillins.<br />

Penicillins, Level 2 (delayed,<br />

Should not be used in same solution as penicillins<br />

Aminoglycosides, parenteral<br />

parenteral moderate, possible)<br />

may inactivate certain aminoglycosides.<br />

Penicillins, Level 2 (delayed, major,<br />

IV doses of penicillins can increase bleeding by<br />

Anticoagulants<br />

parenteral suspected)<br />

prolonging bleeding times.<br />

Penicillins, oral<br />

Level 2 (rapid,<br />

moderate, suspected)<br />

Beta blockers<br />

Ampicillin may reduce the bioavailablility of atenolol.<br />

Some reports of anaphylactic reactions potentiated by<br />

beta-blockers.<br />

Ampicillin<br />

Level 2 (delayed,<br />

Rate of skin rash is potentiated by concomitant use with<br />

Allopurinol<br />

moderate, suspected)<br />

allopurinol<br />

Penicillin G<br />

Level 2 (delayed, minor,<br />

These drugs compete with penicillin G for renal tubular<br />

Diuretics, aspirin, sulfonamides<br />

suspected)<br />

secretion and may prolong penicillin half-life.<br />

Penicillins<br />

Level 4 (delayed,<br />

Oral contraceptives<br />

Oral contraceptive efficacy may be reduced and<br />

Penicillins,<br />

parenteral<br />

Nafcillin<br />

Penicillins<br />

Penicillins<br />

moderate, possible)<br />

Level 4 (rapid,<br />

moderate, possible)<br />

Level 4 (delayed, major,<br />

possible)<br />

Level 4 (delayed,<br />

moderate, possible<br />

Level 5 (delayed, minor,<br />

possible)<br />

Heparin<br />

Cyclosporin<br />

Chloramphenicol<br />

Erythromycin<br />

increased bleeding may occur.<br />

Increased risk of bleeding may occur.<br />

Nafcillin may cause subtherapeutic cyclosporine levels.<br />

Antagonistic effects reported in animal studies.<br />

Clinical studies have demonstrated both antagonistic<br />

and synergistic effects.<br />

Other interactions (per manufacturers labeling): 5-6<br />

• Penicillins and NSAIDs: Since NSAIDs are highly protein bound; they can theoretically<br />

be displaced from binding sites by penicillins.<br />

• Penicillins and Colestipol: Serum concentrations of penicillins are reportedly decreased if<br />

Colestipol is given simultaneously.<br />

179


• Penicillins (e.g., amoxicillin, carbenicillin, mezlocillin) and Methotrexate: Concomitant<br />

use of these agents may decrease the clearance of methotrexate, but inhibiting renal<br />

tubular secretion of the drug.<br />

• Ticarcillin and lithium: Because of high sodium content in commercially available<br />

ticarcillin, lithium’s renal elimination can be displaced resulting in toxicity. Patients<br />

receiving lithium with extended-spectrum penicillins should be closely monitored.<br />

VI.<br />

Adverse Drug Events of the Single Entity Penicillins<br />

Table 6. Common Adverse Events Overview for Single Entity Penicillins 4-6<br />

Ticarcillin<br />

Piperacillin<br />

Carbenicillin<br />

Ampicillin<br />

Amoxicillin<br />

Oxacillin<br />

Nafcillin<br />

Dicloxacillin<br />

Penicillin G, V<br />

Local<br />

Phlebitis + - ++ + - - - 4% 3%<br />

Hypersensitivity<br />

Fever<br />

Rash<br />

Photosensitivity<br />

Anaphylaxis<br />

Serum Sickness<br />

Hematologic<br />

Neutropenia<br />

Eosinophilia<br />

Thrombocytopenia<br />

↑ PT/PTT<br />

+<br />

3%<br />

-<br />

R<br />

4%<br />

R<br />

+<br />

R<br />

R<br />

+<br />

4%<br />

-<br />

-<br />

-<br />

-<br />

+<br />

-<br />

-<br />

+<br />

4%<br />

-<br />

R<br />

-<br />

+<br />

22%<br />

R<br />

+<br />

+<br />

4%<br />

-<br />

R<br />

R<br />

22%<br />

R<br />

-<br />

+<br />

5%<br />

-<br />

-<br />

-<br />

+<br />

2%<br />

R<br />

+<br />

+<br />

5%<br />

-<br />

R<br />

-<br />

+<br />

22%<br />

R<br />

+<br />

+<br />

-<br />

-<br />

+<br />

-<br />

+<br />

+<br />

+<br />

+<br />

+<br />

1%<br />

-<br />

-<br />

-<br />

6%<br />

+<br />

+<br />

+<br />

+<br />

3%<br />

-<br />

+<br />

+<br />

-<br />

+<br />

R<br />

+<br />

GI<br />

Nausea/Vomiting<br />

Diarrhea<br />

C. difficile colitis<br />

-<br />

-<br />

-<br />

+<br />

+<br />

R<br />

-<br />

-<br />

R<br />

-<br />

-<br />

R<br />

2%<br />

5%<br />

R<br />

2%<br />

10%<br />

R<br />

+<br />

+<br />

R<br />

+<br />

2%<br />

+<br />

+<br />

3%<br />

+<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatic failure<br />

CNS<br />

Headache<br />

Confusion<br />

Seizures<br />

Special Senses<br />

Ototoxicity<br />

Vestibular<br />

R<br />

-<br />

R<br />

R<br />

R<br />

-<br />

-<br />

R<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

Renal<br />

↑ BUN, Cr R - - - R R + - -<br />

Cardiac<br />

Dysrhythmias R - - - - - - - -<br />

+ = Occurs, incidence not available; ++ = Significant adverse reaction; R = Rare, defined as < 1%<br />

-<br />

-<br />

R<br />

R<br />

-<br />

-<br />

-<br />

+<br />

-<br />

R<br />

R<br />

+<br />

-<br />

-<br />

R<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

R<br />

-<br />

R<br />

R<br />

R<br />

-<br />

-<br />

+<br />

-<br />

+<br />

-<br />

+<br />

-<br />

-<br />

+<br />

-<br />

R<br />

R<br />

-<br />

-<br />

-<br />

-<br />

-<br />

R<br />

R<br />

R<br />

-<br />

-<br />

180


VII.<br />

Dosing and Administration for the Single Entity Penicillins<br />

Table 7. Dosing for the Single Entity Penicillins 4-6<br />

Drug Usual Adult Dose Comments, Adverse-Reactions<br />

Natural Penicillins<br />

Benzathine Pen G<br />

600,000-1.2 million u IM Q -4<br />

weeks<br />

Hoignes syndrome--An immediate, but transient<br />

neurological reaction with bizarre behavior can occur from<br />

the use of benzathine Pen G or procaine Pen G.<br />

Penicillin G<br />

Low: 600,000-1.2 million u/d IM<br />

High: > 20million u QD IV<br />

Penicillin V<br />

250-500mg BID, TID, QID before<br />

meals and bedtime<br />

Penicillinase-Resistant Penicillins<br />

Dicloxacillin<br />

125-500mg PO Q6h AC<br />

Acute hemorrhagic cystitis reported from high blood levels.<br />

Nafcillin<br />

Oxacillin<br />

1-2g IV/IM Q4h<br />

1-2g IV/IM Q4h<br />

Extravasations can result in tissue necrosis.<br />

Reversible neutropenia (over 10% with >21 day therapy).<br />

Hepatic dysfunction with greater than 12g/day.<br />

Aminopenicillins<br />

Amoxicillin<br />

Ampicillin<br />

500mg PO Q8h or 875mg Q12h<br />

250-500mg PO Q6h<br />

A maculopapular rash occurs, but not a true PCN allergy.<br />

Extended-Spectrum Penicillins<br />

Mezlocillin<br />

3g IV Q4h<br />

1.85mEq Na/g<br />

Piperacillin<br />

Ticarcillin<br />

3-4g IV Q4-6h<br />

UTI: 2g IV Q6h<br />

3g IV Q4-6h<br />

Interferes with platelet function, may prolong bleeding<br />

times.<br />

Special Dosing Considerations<br />

Table 8. Special Dosing Considerations for the Single Entity Penicillins 4-6<br />

Drug Half-life (Normal/ESRD) hr Adjusted for Renal Failure<br />

Estimated CrCl ml/min<br />

Natural Penicillins >50-90 10-50


Aminopenicillins<br />

Amoxicillin/Ampicillin<br />

1.0/5-20: 1.0/7-20<br />

q8h/q6h<br />

q8-12h/q6-12h<br />

q24h/q12-24h<br />

Amoxicillin/clavulanate<br />

1.3/1.0:5-20/4.0<br />

500/125mg q8h<br />

250-500mg AM<br />

component q12h<br />

250-500mg AM<br />

component q24h<br />

Ampicillin/sulbactam<br />

1.0/1.0:9.0/10.0<br />

q6h<br />

q8-12h<br />

q24h<br />

Extended-Spectrum Penicillins<br />

Mezlocillin<br />

1.1/2.6-5.4<br />

q4-6h<br />

q6-8h<br />

q8h<br />

Piperacillin<br />

1.0/3.3-5.1<br />

q4-6h<br />

q6-8h<br />

q8h<br />

Piperacillin/tazobactam<br />

1.0(P)/1.0(T)/3.0(P)4.0(T)<br />

3.375g q6h<br />

2.25g q6h<br />

2.25g q8h<br />

Ticarcillin<br />

1.2/13<br />

1-2g q4h<br />

1-2g q8h<br />

1-2g q12h<br />

Ticarcillin/clavulanate<br />

1.0(T)/1.0(C)/13(T)4(C)<br />

3.1g q4h<br />

2.0g q4-6h<br />

2.0g q12h<br />

182


VIII. Comparative Effectiveness of the Single Entity Penicillins<br />

Table 8 describes the clinical studies for the single entity penicillin.<br />

Table 9. Outcomes Evidence for the Single Entity Penicillin Antibiotics<br />

Study Sample Design Results<br />

High-dose vs. standarddose<br />

-- 3 year, randomized,<br />

acute otitis media 7 amoxicillin for<br />

double-blind<br />

trial<br />

Amoxicillin vs.<br />

azithromycin for C.<br />

trachomatis in<br />

pregnancy before 33<br />

weeks gestation 8 n=39 Randomized,<br />

single-blind trial<br />

Single additional dose n=292 Randomized,<br />

chorioamnionitis 9<br />

of ampicillin plus<br />

controlled trial<br />

gentamicin vs.<br />

continued therapy in<br />

Ampicillin vs. cefotetan<br />

vs. ampicillin/sulbactam<br />

for prevention of post-<br />

Cesarean<br />

endomyometritis 10 n=298 Prospective,<br />

randomized,<br />

double-blind trial<br />

Nafcillin vs.<br />

vancomycin for<br />

staphylococcal<br />

endocarditis 11 n=56 Retrospective<br />

review<br />

Oxacillin/dicloxacillin<br />

vs. linezolid for<br />

complicated skin and<br />

soft tissue infections 12 n=819 Multicenter,<br />

randomized,<br />

double-blind trial<br />

Oxacillin or<br />

vancomycin plus<br />

gentamicin vs.<br />

ciprofloxacin plus<br />

rifampin for<br />

staphylococcal<br />

endocarditis in injection<br />

drug users 13 n=85 Prospective,<br />

randomized, nonblinded<br />

trial<br />

Pediatric patients received either high-dose (80 to<br />

90mg/kg/day) or standard-dose (40 to 45mg/kg/day)<br />

for 3 to 4 days.<br />

• Absolute difference of antibiotic failure rate<br />

between the two groups was nonsignificant<br />

(95% CI, -1.5 to 3.4%; p=0.78).<br />

• Mean duration of illness was similar (3 ± 2<br />

days).<br />

Patients received amoxicillin 500mg TID for 7 days<br />

or a single dose of azithromycin 1g.<br />

• No statistically significant differences in<br />

side effects, compliance, or efficacy.<br />

Patients received ampicillin 2g every 6 hours and<br />

gentamicin 1.5mg/kg every 8 hours when<br />

chorioamnionitis was diagnosed. After delivery, they<br />

received either only 1 additional dose or continued<br />

therapy until afebrile and asymptomatic for 24 hours.<br />

• Failure rate was 3.5% in the continued<br />

therapy group vs. 4.6% in the single<br />

additional dose group (p=0.639).<br />

All patients received single-dose antibiotic<br />

prophylaxis following umbilical cord clamping.<br />

• Frequency of endomyometritis was 4% in<br />

the ampicillin/sulbactam group, 4.2% in the<br />

cefotetan group, and 5.9% in the ampicillin<br />

group.<br />

In patients with methicillin-sensitive Staphylococcus<br />

aureus infection:<br />

• Complete response rate was 74% in the<br />

nafcillin group vs. 50% in the vancomycin<br />

group (p=0.12).<br />

• Mortality rate was 22% in the nafcillin<br />

group vs. 28% in the vancomycin group<br />

(p=0.73).<br />

Patients received oxacillin 2g IV every 6 hours<br />

followed by dicloxacillin 500mg PO every 6 hours or<br />

linezolid 600mg IV every 12 hours.<br />

• Clinical cure rate was 64.9% in the<br />

oxacillin/dicloxacillin group vs. 69.8% in<br />

the linezolid group (p=0.141).<br />

Patients received ciprofloxacin 750mg BID plus<br />

rifampin 300mg BID (oral arm), or oxacillin 2g every<br />

4 hours or vancomycin 1g every 12 hours plus<br />

gentamicin 2mg/kg every 8 hours for the first 5 days<br />

(IV arm). They received 28 days of therapy.<br />

• Observed cure rate was 95% in the oral arm<br />

and 88% in the IV arm (p=0.6).<br />

• Drug toxicity was observed in 2.8% of the


oral arm and in 61.5% of the IV arm<br />

(p


IX.<br />

Conclusions<br />

The single entity penicillins have been useful antibiotics for many types of infections, for many<br />

years. Although resistance is an issue with some drugs in this class, the penicillin antibiotics<br />

remain safe and effective for the first-line treatment of many bacterial organisms in adults and<br />

children. All oral, single entity natural penicillins, penicillinase-resistant penicillins, and<br />

aminopenicillins in this class are available in generic formulations and are available to recipients.<br />

The extended spectrum penicillins (one oral agent and two injectables) may have a use for more<br />

serious infections requiring hospitalization, but their use is not applicable to the general<br />

population. The extended spectrum penicillins should be available for special<br />

needs/circumstances that require medical justification through the prior authorization program.<br />

Therefore, all brand products within the class are comparable to each other and to the generics and<br />

OTC products in the class and offer no significant clinical advantage over other alternatives in<br />

general use.<br />

X. Recommendations<br />

No brand single entity penicillin is recommended for preferred status.<br />

185


I. Overview<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Penicillin Antibiotics<br />

Combination Agents<br />

AHFS 081216<br />

Several penicillin agents such as amoxicillin, ampicillin, piperacillin and ticarcillin are available in<br />

fixed combinations with ß-lactam inhibitors (Clavulanic acid, Sulbactam, Tazobactam). These<br />

combinations act synergistically to expand the spectrum of activity against many strains of ß-<br />

lactamase producing microorganisms. 1<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this review.<br />

Table 1. Combination Penicillin Antibiotics in this <strong>Review</strong> 2<br />

Generic Name Formulation Example Brand Name<br />

Amoxicillin/Clavulanate Suspension, Tablets,<br />

Chewable Tablets, Filmcoated<br />

Augmentin*, Augmentin<br />

ES-600, Augmentin XR<br />

Tablets<br />

Ampicillin/Sulbactam Injection (IV, IM) Unasyn*<br />

Piperacillin/Tazobactam Injection (IV) Zosyn<br />

Ticarcillin/Clavulanate Injection (IV) Timentin<br />

*Generic Available.<br />

Table 2. Comparison of Bacterial Coverage of the Combination Penicillins 2<br />

Gram-Positive<br />

Amoxicillin/ Ampicillin/ Piperacillin/<br />

✔ = Generally Susceptible<br />

Clavulanate Sulbactam Tazobactam<br />

Staphylococci<br />

Staphylococcus aureus<br />

Staphylococcus<br />

epidermidis<br />

Streptococci<br />

Streptococcus<br />

Pneumoniae<br />

Beta-hemolytic streptococci<br />

Enterococcus faecalis<br />

Streptococcus viridans<br />

Corynebaacterium<br />

diphtheriae<br />

Bacillus anthracis<br />

Streptococcus agalactiae<br />

Streptococcus pyogens<br />

Ticarcillin/<br />

Clavulanate<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔<br />

✔<br />

Gram-<br />

Negative<br />

Erysipelothrix<br />

rhusiopathiae<br />

Listeria monocytogenes<br />

Escherichia coli<br />

Haemophilus influenzae<br />

Eikenella corrodens<br />

Bacteroides<br />

melaninogenicus<br />

✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔<br />

186


Anaerobic<br />

Klebsiella sp.<br />

Neisseria gonorrhoeae<br />

Neisseria meningitides<br />

Proteus mirabilis<br />

Salmonella sp.<br />

Shigella sp.<br />

Morganella morganii<br />

Proteus vulgaris<br />

Providencia stuartii<br />

Enterobacter sp.<br />

Citrobacter sp.<br />

Pseudomonas aeruginosa<br />

Serratia sp.<br />

Acinetobacter sp.<br />

Streptobacillus<br />

moniliformis<br />

✔ = Generally<br />

Susceptible<br />

Clostridium sp.<br />

Peptococcus sp.<br />

Peptostreptococcus sp.<br />

Bacteroides sp.<br />

Fusobacterium sp.<br />

Eubacterium sp.<br />

Treponema pallidum<br />

Actinomyces bovis<br />

Veillonella sp.<br />

✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔<br />

✔<br />

✔ ✔ ✔<br />

✔ ✔ ✔<br />

✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔<br />

Amoxicillin/<br />

Clavulanate<br />

✔<br />

✔<br />

Ampicillin/<br />

Sulbactam<br />

✔<br />

✔<br />

✔<br />

Piperacillin/<br />

Tazobactam<br />

✔<br />

✔<br />

✔<br />

✔<br />

Ticarcillin/<br />

Clavulanate<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔<br />

✔ ✔ ✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

✔<br />

187


II.<br />

Indications of the Combination Penicillin Antibiotics<br />

Table 3. FDA-Approved Indications for the Combination Penicillins 2<br />

Drug<br />

UTI<br />

Lower RTI<br />

Intraabdominal<br />

Infections<br />

Gynecologic<br />

Infections<br />

Septicemia<br />

Bone and Joint<br />

Infections<br />

Skin and Skin<br />

Structure<br />

Infections<br />

Endometriosis/<br />

PID<br />

CAP<br />

Nosocomial<br />

Pneumonia<br />

Otitis Media/<br />

Sinusitis<br />

Appendicitis<br />

Amoxicillin/<br />

Clavulanate<br />

Ampicillin/<br />

Sulbactam<br />

Piperacillin/<br />

Tazobactam<br />

Ticarcillin/<br />

Clavulanate<br />

* Augmentin XR<br />

✔ ✔ ✔ ✔ ✔ ✔ * ✔<br />

✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔<br />

✔ ✔ ✔ ✔ ✔ ✔ ✔<br />

III.<br />

Pharmacokinetic Parameters of the Combination Penicillins<br />

Table 4. Pharmacokinetic Parameters of the Combination Penicillins 1-5<br />

Drug Mechanism of Bioavailability Protein Metabolism Elimination Half-Life<br />

Action<br />

Binding<br />

Amoxicillin/<br />

Clavulanate<br />

Ampicillin/<br />

Sulbactam<br />

Piperacillin/<br />

Tazobactam<br />

Amoxicillin<br />

inhibits cell wall<br />

synthesis by<br />

binding to specific<br />

penicillin-binding<br />

proteins (PBPs)<br />

located inside the<br />

bacterial cell wall.<br />

Clavulanic<br />

inhibits β-<br />

lactamases that<br />

inactivate<br />

penicillins<br />

Ampicillin inhibits<br />

cell wall synthesis<br />

by binding to<br />

specific penicillinbinding<br />

proteins<br />

(PBPs) located<br />

inside the bacterial<br />

cell wall.<br />

Sulbactam<br />

inhibits β-<br />

lactamases that<br />

inactivate<br />

penicillins<br />

Piperacillin<br />

inhibits cell wall<br />

synthesis by<br />

binding to specific<br />

penicillin-binding<br />

75%<br />

Peak<br />

concentrations<br />

occur in 1-2.5<br />

hours<br />

Peak serum<br />

concentrations<br />

are obtained<br />

immediately<br />

following IV<br />

infusion. Peak<br />

serum<br />

concentrations<br />

when given<br />

IM are<br />

obtained<br />

within 30-40<br />

minutes.<br />

Both<br />

bioavailability<br />

and peak<br />

concentrations<br />

are dose<br />

17%<br />

amoxicillin<br />

and 25%<br />

clavulanic<br />

Ampicillin:<br />

28%<br />

Sulbactam:<br />

38%<br />

Piperacillin:<br />

26%-33%<br />

Tazobactam<br />

: 31%-32%<br />

188<br />

Clavulanic<br />

acid is<br />

metabolized<br />

hepatically<br />

Piperacillin:<br />

6%-9%,<br />

Tazobactam:<br />

26%<br />

80% of a dose<br />

of amoxicillin is<br />

excreted<br />

unchanged in<br />

urine;<br />

clavulanic 30%-<br />

40%<br />

- 75%-80% of<br />

both drugs are<br />

excreted<br />

unchanged in<br />

the urine within<br />

8 hours after<br />

administration<br />

Piperacillin:<br />

50%-70%<br />

unchanged in<br />

the urine, 10%-<br />

20% eliminated<br />

1.2 hours in<br />

normal renal<br />

function<br />

Ampicillin:<br />

1-1.8 hours<br />

Sulbactam:<br />

1-1.3 hours<br />

in patients<br />

with normal<br />

renal<br />

function.<br />

Piperacillin:<br />

1hour,<br />

metabolite:<br />

1-1.5 hours.<br />

Tazobactam:


Ticarcillin/<br />

Clavulanate<br />

proteins (PBPs)<br />

located inside the<br />

bacterial cell wall.<br />

Tazobactam<br />

inhibits β-<br />

lactamases that<br />

inactivate<br />

penicillins<br />

Ticarcillin inhibits<br />

bacterial cell wall<br />

synthesis by<br />

binding to specific<br />

penicillin-binding<br />

proteins (PBPs)<br />

located inside the<br />

bacterial cell wall.<br />

Clavulanic<br />

inhibits β-<br />

lactamases that<br />

inactivate<br />

penicillins<br />

proportional<br />

Peak serum<br />

concentrations<br />

are obtained<br />

immediately<br />

following IV<br />

infusion.<br />

Ticarcillin:<br />

45%-65%<br />

Clavulanic:<br />

9%-30%<br />

Clavulanic<br />

acid is<br />

metabolized<br />

hepatically<br />

in bile.<br />

Tazobactam:<br />

found in the<br />

urine in 24<br />

hours, with<br />

26% as active<br />

metabolite<br />

45% excreted<br />

unchanged in<br />

the urine. 60%-<br />

90% of<br />

ticarcillin<br />

excreted<br />

unchanged in<br />

the urine<br />

0.7-0.9<br />

hours<br />

Clavulanate:<br />

66-90<br />

minutes,<br />

Ticarcillin:<br />

66-72<br />

minutes in<br />

patients with<br />

normal renal<br />

function.<br />

IV.<br />

Drug Interactions of the Combination Penicillins<br />

Table 5 lists the most significant drug drug-interactions (Level 1 and 2) for the drugs indexed by<br />

Drug Interactions Facts. 2,6<br />

Table 5. Drug Interactions of the Combination Penicillins 2,6<br />

Drug Significance Interaction Mechanism<br />

Penicillins<br />

Level 1<br />

Tetracyclines impair the bacteriocidal effects<br />

Tetracyclines<br />

of penicillins.<br />

Level 2<br />

Should not be used in same solution as<br />

Penicillins, parenteral<br />

Aminoglycosides, parenteral penicillins may inactivate certain<br />

aminoglycosides.<br />

Penicillins, parenteral<br />

Level 2<br />

IV doses of penicillins can increase bleeding<br />

Anticoagulants<br />

by prolonging bleeding times.<br />

Level 2<br />

Ampicillin may reduce the bioavailability of<br />

Penicillins, oral<br />

Beta blockers atenolol. Some reports of anaphylactic<br />

reactions potentiated by beta-blockers.<br />

Ampicillin<br />

Level 2<br />

Rate of skin rash is potentiated by<br />

Allopurinol<br />

concomitant use with allopurinol<br />

Table 6. Other Reported Drug Interactions of the Combination Penicillins 6<br />

Precipitant<br />

Drug<br />

Parenteral<br />

penicillins<br />

Object Drug *<br />

Heparin<br />

Penicillin Drug Interactions<br />

Description<br />

Heparin with high-dose penicillins may increase risk of bleeding.<br />

Probenecid Penicillin Coadministration results in increases in penicillin levels and half-life; do not give<br />

probenecid concurrently.<br />

Penicillin<br />

Neuromuscular<br />

blockers<br />

Increase duration of blockade<br />

*<br />

= Object drug increased.<br />

189


V. Adverse Events of the Combination Penicillins Antibiotics<br />

Table 7. Common Adverse Events (%) Reported for the Combination Penicllins 4<br />

Amoxicillin/<br />

Clavulanate<br />

Ampicillin/<br />

Sulbactam<br />

Piperacillin/<br />

Tazobactam<br />

Ticarcillin/<br />

Clavulanate<br />

Local<br />

Phlebitis - 3% 1% -<br />

Hypersensitivity<br />

Fever<br />

Rash<br />

Photosensitivity<br />

Anaphylaxis<br />

Serum Sickness<br />

Hematologic<br />

Neutropenia<br />

Eosinophilia<br />

Thrombocytopenia<br />

↑ PT/PTT<br />

+<br />

3%<br />

-<br />

R<br />

-<br />

+<br />

+<br />

R<br />

-<br />

+<br />

2%<br />

-<br />

-<br />

+<br />

+<br />

22%<br />

R<br />

-<br />

2%<br />

4%<br />

-<br />

-<br />

+<br />

+<br />

+<br />

+<br />

+<br />

+<br />

2%<br />

-<br />

+<br />

+<br />

+<br />

5%<br />

R<br />

+<br />

GI<br />

Nausea/Vomiting<br />

Diarrhea<br />

C. difficile colitis<br />

3%<br />

9%<br />

+<br />

+<br />

2%<br />

+<br />

7%<br />

11%<br />

+<br />

1%<br />

1%<br />

+<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatic failure<br />

CNS<br />

Headache<br />

Confusion<br />

Seizures<br />

Special Senses<br />

Ototoxicity<br />

Vestibular<br />

+<br />

-<br />

+<br />

-<br />

R<br />

-<br />

-<br />

Renal<br />

↑ BUN, Cr - R - -<br />

Cardiac<br />

Dysrhythmias - - - -<br />

+ = Occurs, incidence not available; ++ = Significant adverse reaction; R = Rare, defined as < 1%<br />

6%<br />

-<br />

R<br />

R<br />

-<br />

-<br />

-<br />

+<br />

-<br />

8%<br />

R<br />

R<br />

-<br />

-<br />

+<br />

-<br />

R<br />

R<br />

+<br />

-<br />

-<br />

190


VI.<br />

Dosage and Administration for the Combination Penicillin Antibiotics<br />

Table 8. Dosing for the Combination Penicillin Antibiotics 2,5<br />

Drug Availability Dose/Frequency/Duration<br />

Amoxicillin/<br />

clavulanate<br />

Tablets: 250mg/125mg,<br />

500mg /125mg, 875mg/125mg.<br />

Adults:<br />

One 500mg tablet Q12h or 250mg tablet Q8h<br />

Extended Release tablets:<br />

1000mg/62.5mg (scored and<br />

unscored tablets)<br />

Chewable Tablets:<br />

125mg/31.25mg,<br />

250mg/28.5mg, 400mg/57mg.<br />

Oral Suspension (per 5ml):<br />

125mg/31.25mg,<br />

200mg/28.5mg, 250mg/62.5mg,<br />

400mg/57mg, 600mg/42.9mg<br />

(Augmentin ES-600)<br />

Suspension:<br />

125mg/5ml or 250mg/5ml in place of 500mg tablet or<br />

200mg/5ml or 400mg/5ml suspension in place of<br />

875mg<br />

tablet.<br />

Severe infections and respiratory tract infections:<br />

One 875mg tablet every 12 hours or 500mg tablet every 8 hours<br />

Children:<br />

Less than 3 months of age:<br />

30mg/kg/day divided every 12 hours, based on amoxicillin<br />

component.<br />

3 months of age or older:<br />

Refer to table below. 250mg table should not be used until<br />

child weighs 40kg or more<br />

40kg or more:<br />

Dose according to adult recommendations<br />

Amoxicillin/Clavulanate Dosing Regimen in Children > 3 months<br />

of age<br />

Dosing Regimen<br />

Infection<br />

200mg/5ml or<br />

400mg/5ml (Q 12h)<br />

OM 1 , sinusitis, 45mg/kg/day<br />

LRTI 2 , severe<br />

infections<br />

Less severe 25mg/kg/day<br />

infections<br />

1<br />

Otitis Media<br />

2<br />

Lower Respiratory Tract Infection<br />

125mg/5ml or<br />

250mg/5ml (Q 8h)<br />

40mg/kg/day<br />

20mg/kg/day<br />

Augmentin ES-600mg<br />

Augmentin ES-600mg (Pediatric patients 3 months and older)<br />

Body Weight (kg)<br />

Volume of Augmentin ES-600<br />

8 90mg/kg/day<br />

12 3ml twice daily<br />

16 4.5mg twice daily<br />

20 6ml twice daily<br />

24 7.5mg twice daily<br />

28 9ml twice daily<br />

32 10.5ml twice daily<br />

36 13.5ml twice daily<br />

Pediatric patients weighing 40kg or more:<br />

Experience with Augmentin ES-600 not available<br />

191


Augmentin XR:<br />

Augmentin XR Dosing<br />

Indication Dose Duration<br />

Sinusitis 2 tablets Q 12h 10 days<br />

Communityacquired<br />

2 tablets Q 12h 7 – 10 days<br />

Pneumonia<br />

Ampicillin/<br />

Sulbactam<br />

Piperacillin/<br />

Tazobactam<br />

Ticarcillin/<br />

Clavulanate<br />

Powder for Injection:<br />

1.5g (1g ampicillin/0.5g<br />

sulbactam in vials and ADD-<br />

Vantage vials)<br />

3g (2g ampicillin/1g sulbactam<br />

in vials and ADD-Vantage<br />

vials)<br />

15g (10g ampicillin/5g<br />

sulbactam in bulk package)<br />

Powder for Injection<br />

(lyophilized):<br />

2.25g (2g piperacillin/0.25g<br />

tazobactam in vials and ADD-<br />

Vantage vials-4.69mEq sodium)<br />

3.375g (3g piperacillin/0.375mg<br />

tazobactam- 7.04mEq sodium)<br />

4.5g (4g piperacillin/0.5g<br />

tazobactam- 9.39mEq sodium)<br />

40.5g (36g piperacillin/4.5g<br />

tazobactam- 84.5mEq sodium)<br />

Injection:<br />

2.25g/50ml (2g<br />

piperacillin/0.25g tazobactam-<br />

5.7mEq sodium)<br />

3.375g/50ml (3g<br />

piperacillin/0.375g tazobactam<br />

8.6mEq sodium)<br />

4.5g/100ml (4g piperacillin/0.5g<br />

tazobactam- 11.4mEq sodium)<br />

Powder for Injection:<br />

3g ticarcillin and 0.1g clavulanic<br />

acid. In piggyback bottles,<br />

ADD-Vantage vials and 31g<br />

bulk packages<br />

Injection, solution:<br />

3g ticarcillin and 0.1g clavulanic<br />

acid. In 100ml premixed, frozen<br />

Galaxy plastic containers.<br />

Given IV or IM<br />

Adults:<br />

1.5mg – 3mg every 6 hours. Do not exceed 4g/day<br />

Children 40kg or more:<br />

Dose according to adult recommendations<br />

Adults:<br />

Administer by IV infusion over 30 minutes. The usual total<br />

daily dose for adults is 3.375g every 6 hours totaling 13.5g for<br />

7-10 days.<br />

Nosocomial Pneumonia:<br />

Start with 4.5g every 6 hours plus an aminoglycoside. The<br />

recommended total daily dose is 18g for 7-14 days. Continue<br />

aminoglycoside in patients where P. aeruginosa is isolated.<br />

Administer by IV infusion over 30minutes. Usual duration is<br />

approximately 10-14 days.<br />

Adults:<br />

Systemic and Gynecological Infections<br />

Urinary<br />

Infections<br />

Moderate Severe<br />

Adults > 60kg 3.1g every 4-6h 200mg/kg/day<br />

every 6hrs<br />

Adults < 60kg<br />

300mg/kg/<br />

day every<br />

4hrs<br />

200-300mg/kg/day every 4-6 hours<br />

192


Children:<br />

Dosing Guidelines for Children > 3 Months of age<br />

Mild to Moderate<br />

Infection<br />

Severe Infections<br />

> 60kg 3.1g every 6h 3.1g every 4h<br />

< 60kg 200-300mg/kg/day<br />

every 6h<br />

300mg/kg/day every<br />

4h<br />

Special Dosing Considerations<br />

Table 9. Special Dosing considerations for the Combination Penicillins 2,5<br />

Drug Renal<br />

Dosing<br />

Hepatic<br />

dosing<br />

Pediatric Use Pregnancy<br />

Category<br />

Amoxicillin/<br />

Doses provided in B<br />

clavulanate<br />

previous section.<br />

Ampicillin/<br />

sulbactam<br />

Piperacillin/<br />

tazobactam<br />

Ticarcillin/<br />

clavulanate<br />

No, unless<br />

severe<br />

impairment<br />

(CrCl


Amoxicillin/clavulanate<br />

plus ciprofloxacin vs.<br />

gentamicin plus<br />

piperacillin/tazobactam<br />

for low-risk febrile<br />

neutropenia 19 n=126 Prospective,<br />

randomized,<br />

controlled trial<br />

Study conducted in<br />

the UK.<br />

Amoxicillin/clavulanate<br />

vs. telithromycin for<br />

acute maxillary<br />

sinusitis 20 n=434 Assessed 17 to 24<br />

days post-therapy,<br />

multicenter,<br />

randomized,<br />

double-blind trial<br />

Pharmacokinetically<br />

enhanced<br />

Amoxicillin/clavulanate<br />

(Augmentin XR) for<br />

respiratory tract<br />

infections caused by S.<br />

pneumoniae 21 n=427 Pooled analysis of<br />

nine clinical<br />

studies<br />

Ampicillin/sulbactam<br />

vs. clindamycin ±<br />

cephalosporin for<br />

aspiration pneumonia<br />

and lung abscess 22 n=70 Open-label,<br />

multicenter,<br />

prospective,<br />

randomized,<br />

comparative trial<br />

Study conducted in<br />

Germany.<br />

Ampicillin/sulbactam<br />

vs. cefuroxime for<br />

serious skin and skin<br />

structure infections in<br />

pediatrics 23 n=98 Open-label,<br />

multicenter,<br />

prospective,<br />

randomized trial<br />

Ciprofloxacin plus<br />

piperacillin vs.<br />

tobramycin plus<br />

piperacillin for empiric<br />

therapy of febrile<br />

neutropenia 24 n=471 Multicenter,<br />

randomized,<br />

double-blind,<br />

controlled trial<br />

194<br />

Patients received amoxicillin/clavulanate 500/175mg<br />

every 8 hours plus ciprofloxacin 750mg BID for 5<br />

days (oral arm) or gentamicin 80mg every 8 hours<br />

plus piperacillin/tazobactam 4.5 g every 8 hours (IV<br />

arm) until hospital discharge.<br />

• Initial treatment was successful without<br />

antibiotic modification in 90% of IV arm<br />

vs. 84.8% in the oral arm (p=0.55).<br />

• Median patient stay was 4 days in the IV<br />

arm vs. 2 days in the oral arm (p


Piperacillin/tazobactam<br />

vs. ertapenem for<br />

complicated skin and<br />

skin structure infections<br />

caused by MSSA 25 n=135 Assessed 10 to 21<br />

days post-therapy,<br />

randomized,<br />

double-blind,<br />

controlled trial<br />

Piperacillin/tazobactam n=316 Assessed 2 to 4<br />

pelvic infections 26 therapy,<br />

vs. ertapenem for acute<br />

weeks post-<br />

multicenter,<br />

randomized,<br />

double-blind,<br />

controlled trial<br />

Piperacillin/tazobactam<br />

vs. ciprofloxacin plus<br />

amoxicillin ±<br />

metronidazole for<br />

infections after liver<br />

transplantation 27 n=217 3 month,<br />

prospective,<br />

randomized trial<br />

Study conducted in<br />

London.<br />

Piperacillin/tazobactam<br />

plus tobramycin vs.<br />

ceftazidime plus<br />

tobramycin for<br />

nosocomial lower<br />

respiratory tract<br />

infection 28 n=136 Open-label,<br />

multicenter,<br />

randomized,<br />

comparative trial<br />

Piperacillin/tazobactam<br />

plus amikacin vs.<br />

ceftazidime plus<br />

amikacin for ventilatorassociated<br />

pneumonia 29 n=115 Open-label,<br />

multicenter,<br />

randomized,<br />

controlled trial<br />

Study conducted in<br />

France.<br />

195<br />

in the tobramycin/piperacillin group (95%<br />

CI, -2.2 to 6.4%).<br />

• Fevers resolved faster in the<br />

ciprofloxacin/piperacillin group vs. the<br />

tobramycin/piperacillin group (5 vs. 6 days,<br />

respectively; p=0.005).<br />

• No significant differences in adverse events<br />

or toxicity (p=0.083).<br />

Patients received ertapenem 1g daily or<br />

piperacillin/tazobactam 3.375g every 6 hours.<br />

• Use of ertapenem resulted in cure rate of<br />

80.6% compared to 80.9% in the<br />

piperacillin/tazobactam group (p=0.99).<br />

Patients received ertapenem 1g daily or<br />

piperacillin/tazobactam 3.375g every 6 hours.<br />

• Median duration of therapy was 4 days.<br />

• Most common pathogen was E. coli.<br />

• Use of ertapenem resulted in a cure rate of<br />

93.9% compared to 91.5% in the<br />

piperacillin/tazobactam group (95% CI, -4<br />

to 8.8%).<br />

Patients received piperacillin/tazobactam or<br />

ciprofloxacin/amoxicillin for the first 3 months after<br />

liver transplant. Patients in the<br />

ciprofloxacin/amoxicillin group also received<br />

metronidazole if anaerobic infection was suspected.<br />

• At 72 hours, overall response rate was<br />

66.1% in the piperacillin/tazobactam group<br />

vs. 60% in the ciprofloxacin/amoxicillin<br />

group (p=0.399).<br />

• At end-of-study, successful clinical<br />

outcome rate was 58.9% in the<br />

piperacillin/tazobactam group vs. 50.5% in<br />

the ciprofloxacin/amoxicillin group<br />

(p=0.222).<br />

Patients received piperacillin/tazobactam 3.375g<br />

every 4 hours or ceftazidime 2g every 8 hours. All<br />

patients received tobramycin 5mg/kg/day divided<br />

every 8 hours.<br />

• Clinical success rate was 74% in the<br />

piperacillin/tazobactam group vs. 50% in<br />

the ceftazidime group (p=0.006).<br />

• Eradication rate of baseline pathogen was<br />

66% in the piperacillin/tazobactam group<br />

vs. 38% in the ceftazidime group (p=0.003).<br />

• Mortality rate in the piperacillin/tazobactam<br />

group was 7.7% vs. 17% in the ceftazidime<br />

group (p=0.03).<br />

Patients received piperacillin/tazobactam 4.5g QID<br />

or ceftazidime 1g QID. All patients received<br />

amikacin 7.5mg/kg BID.<br />

• Bacteriologically confirmed infections<br />

included polymicrobial (37%) and<br />

involving P. aeruginosa (32%).<br />

• Clinical cure rate was 51% with<br />

piperacillin/tazobactam vs. 36% with<br />

ceftazidime (95% CI, -0.2 to 30.2%).


Ticarcillin/clavulanate<br />

plus aztreonam vs.<br />

cefepime for febrile<br />

neutropenia in high-dose<br />

chemotherapy patients 30 n=126 Open-label,<br />

randomized trial<br />

Ticarcillin/clavulanate<br />

vs. Imipenem/cilistatin<br />

for gangrenous and<br />

perforated appendicitis 31 n=137 Randomized,<br />

double-blind,<br />

controlled trial<br />

• Twenty-eight day mortality rate was 16%<br />

with piperacillin/tazobactam vs. 20% with<br />

ceftazidime (p=0.55).<br />

• Bacteriologic failure rate was 33% with<br />

piperacillin/tazobactam vs. 51% with<br />

ceftazidime (p=0.05).<br />

Patients received ticarcillin/clavulanate 3.1g every 6<br />

hours plus aztreonam 1g every 8 hours or cefepime<br />

2g every 8 hours for 72 hours.<br />

• Clinical response rates were similar in the<br />

two groups (55% for cefepime and 61% for<br />

ticarcillin/clavulanate plus aztreonam).<br />

Patients received ticarcillin/clavulanate 3.1g every 6<br />

hours or imipenem/cilistatin 500mg every 6 hours for<br />

3 to 5 days.<br />

• Clinical success rate was 96.9% in the<br />

ticarcillin/clavulanate group vs. 95.9% in<br />

the imipenem/cilastatin group (p=0.99).<br />

• Bacteriologic success rate was 100% in the<br />

ticarcillin/clavulanate group vs. 98.4% in<br />

the imipenem/cilastatin group (p=0.99).<br />

• Adverse event rate was similar in both<br />

groups (p=0.31).<br />

Additional Evidence<br />

Dose Simplification: In most cases, the penicillins in this class are given in divided doses<br />

depending on the infection being treated. Literature search of Medline/PubMed revealed no<br />

clinical trials available comparing the efficacy of amoxicillin/clavulanate dosed three times daily<br />

and the enhanced formulations dosed twice daily.<br />

Stable Therapy: Not applicable.<br />

Impact on Physician Visits: A search of Medline and Ovid did not reveal data pertinent to<br />

medical resource utilization or the duration of hospitalization.<br />

VIII. Conclusion<br />

Amoxicillin/clavulanate is the only combination penicillin antibiotic in this class with use that is<br />

applicable to the general population. It is recommended as a first or second-line agent for several<br />

different infections (acute otitis media, sinusitis, community-acquired pneumonia). A generic<br />

formulation is available for amoxicillin/clavulanate (Augmentin), but generic formulations are not<br />

available for Augmentin XR or Augmentin ES-600. Augmentin XR has limited indications versus<br />

regular Augmentin, with similar efficacy. Additionally, only regular Augmentin is available in<br />

solid and liquid dosage formulations.<br />

The other combination penicillins in this class (ampicillin/sulbactam, piperacillin/tazobactam, and<br />

ticarcillin/clavulanate) are injections for use in more serious infections typically requiring<br />

hospitalization. These drugs should be available for special needs/circumstances that require<br />

medical justification through the prior authorization program.<br />

Therefore, all brand products within the class are comparable to each other and to the generics and<br />

OTC products in the class and offer no significant clinical advantage over other alternatives in<br />

general use.<br />

IX.<br />

Recommendation<br />

No brand combination penicillin antibiotic is recommended for preferred status.<br />

196


References<br />

1. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004<br />

2. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

3. Abate BJ, Barriere SL. Antimicrobial Regimen Selection. In: <strong>Pharmacotherapy</strong>. A<br />

Pathophysiologic Approach, Fifth Edition. Dipiro JT, Talbert RL, Yee GC, et al. Eds. McGraw-<br />

Hill. New York. 2002. Pg. 1817-29.<br />

4. Gilbert DN, Moellering, RC, Sande MA. The Sanford Guide to Antimicrobial Therapy 34 th<br />

Edition. Hyde Park, Vermont. 2004.<br />

5. Murray L, Senior Editor. Package inserts. In: Physicians’ Desk Reference, PDR Edition 58,<br />

2004. Thomson PDR. Montvale, NJ. 2004.<br />

6. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

7. Garrison GD, Sorum PC, Hior W, Miller MM. High-dose versus standard-dose amoxicillin for<br />

acute otitis media. Ann Pharmacother. 2004;38(1):158-60.<br />

8. Kacmar J, Cheh E, Montagno A, Peipert JF. A randomized trial of azithromycin versus<br />

amoxicillin for the treatment of Chlamydia trachomatis in pregnancy. Infect Dis Obstet Gynecol.<br />

2001;9(4):197-202.<br />

9. Edwards RK, Duff P. Single additional dose postpartum therapy for women with<br />

chorioamnionitis. Obstet Gynecol. 2003;102(5 Pt 1):957-61.<br />

10. Spinnato JA, Youkilis B, Cook VD, et al. Antibiotic prophylaxis at Cesarean delivery. J Matern<br />

Fetal Med. 2000;9(6):348-50.<br />

11. Gentry CA, Rodvold KA, Novak RM, Hershow RC, Naderer OJ. Retrospective evaluation of<br />

therapies for Staphylococcus aureus endocarditis. <strong>Pharmacotherapy</strong>. 1997;17(5):990-7.<br />

12. Stevens DL, Smith LG, Bruss JB, et al. Randomized comparison of linezolid (PNU-100766)<br />

versus oxacillin-dicloxacillin for treatment of complicated skin and soft tissue infections.<br />

Antimicrob Agents Chemother. 2000;44(12):3408-13.<br />

13. Heldman AW, Hartert TV, Ray SC, et al. Oral antibiotic treatment of right-sided staphylococcal<br />

endocarditis in injection drug users: prospective randomized comparison with parenteral therapy.<br />

Am J Med. 1996;101(1):68-76.<br />

14. Bland ML, Vermillion ST, Soper DE. Late third-trimester treatment of rectovaginal group B<br />

streptococci with benzathine penicillin G. Am J Obstet Gynecol. 2000;183(2):372-6.<br />

15. Hook EW, Martin DH, Stephens J, Smith BS, Smith K. A randomized, comparative pilot study of<br />

azithromycin versus benzathine penicillin G for treatment of early syphilis. Sex Transm Dis.<br />

2002;29(8):486-90.<br />

16. Smith NH, Musher DM, Huang DB, et al. Response of HIV-infected patients with asymptomatic<br />

syphilis to intensive intramuscular therapy with ceftriaxone or procaine penicillin. Int J STD<br />

AIDS. 2004;15(5):328-32.<br />

17. Curtin-Wirt C, Casey JR, Murray PC, et al. Efficacy of penicillin vs. amoxicillin in children with<br />

group A beta hemolytic streptococcal tonsillopharyngitis. Clin Pediatr (Phila). 2003;42(3):219-<br />

25.<br />

18. Lode H, Magyar P, Muir JF, et al. Once-daily oral gatifloxacin vs. three-times-daily co-amoxiclav<br />

in the treatment of patients with community-acquired pneumonia. ClinMicrobiol Infect.<br />

2004;10(6):512-20.<br />

19. Innes HE, Smith DB, O’Reilly SM, et al. Oral antibiotics with early hospital discharge compared<br />

with in-patient intravenous antibiotics for low-risk febrile neutropenia in patients with cancer: a<br />

prospective randomized controlled single centre study. Br J Cancer. 2003;89(1):43-9.<br />

20. Luterman M, Tellier G, Lasko B, Leroy B. Efficacy and tolerability of telithromycin for 5 or 10<br />

days vs. amoxicillin/clavulanic acid for 10 days in acute maxillary sinusitis. Ear Nose Throat J.<br />

2003;82(8):576-80.<br />

21. File TM Jr., Jacobs MR, Poole MD, et al. Outcome of treatment of respiratory tract infections due<br />

to Streptococcus pneumoniae, including drug-resistant strains, with pharmacokinetically enhanced<br />

amoxicillin/clavulanate. Int J Antimicrob Agents. 2002;20(4):235-47.<br />

22. Allewelt M, Schuler P, Bolcskei PL, Mauch H, Lode H, Study Group on Aspiration Pneumonia.<br />

Ampicillin + sulbactam vs. clindamycin ± cephalosporin for the treatment of aspiration pneumonia<br />

and primary lung abscess. Clin Microbiol Infect. 2004;10(2):163-70.<br />

197


23. Azimi PH, Barson WJ, Janner D, Swanson R, UNASYN Pediatric Study Group. Efficacy and<br />

safety of ampicillin/sulbactam and cefuroxime in the treatment of serious skin and skin structure<br />

infections in pediatric patients.<br />

24. Peacock JE, Herrington DA, Wade JC, et al. Ciprofloxacin plus piperacillin compared with<br />

tobramycin plus piperacillin as empirical therapy in febril neutropenic patients. A randomized,<br />

double-blind trial. Ann Intern Med. 2002;137(2):77-87.<br />

25. Gesser RM, McCarroll KA, Woods GL. Efficacy of ertapenem against methicillin-susceptible<br />

Staphylococcus aureus in complicated skin/skin structure infections: results of a double-blind<br />

clinical trial versus piperacillin-tazobactam. Int J Antimicrob Agents. 2004;23(3):235-9.<br />

26. Roy S, Higareda I, Angel-Muller E, Protocol 023 Study Group. Ertapenem once a day versus<br />

piperacillin-tazobactam every 6 hours for treatment of acute pelvic infections: a prospective,<br />

multicenter, randomized, double-blind study. Infect Dis Obstet Gynecol. 2003;11(1):27-37.<br />

27. Philpott-Howard J, Burroughs A, Fisher N, et al. Piperacillin-tazobactam versus ciprofloxacin<br />

plus amoxicillin in the treatment of infective episodes after liver transplantation. J Antimicrob<br />

Chemother. 2003;52(6):993-1000.<br />

28. Joshi M, Bernstein J, Solomkin J, Wester BA, Kuye O, Piperacillin/tazobactam Nosocomial<br />

Pneumonia Study Group. Piperacillin/tazobactam plus tobramycin versus ceftazidime plus<br />

tobramycin for the treatment of patients with nosocomial lower respiratory tract infection. J<br />

Antimicrob Chemother. 1999;43(3):389-97.<br />

29. Brun-Buisson C, Sollet JP, Schweich H, Briere S, Petit C, VAP Study Group. Treatment of<br />

ventilator-associated pneumonia with piperacillin-tazobactam/amikacin versus<br />

ceftazidime/amikacin: a multicenter, randomized controlled trial. Clin Infect Dis.<br />

1998;26(2):346-54.<br />

30. Fleming DR, Ziegler C, Baize T, Mudd L, Goldsmith GH, Herzig RH. Cefepime versus ticarcillin<br />

and clavulanate potassium and aztreonam for febrile neutropenia therapy in high-dose<br />

chemotherapy patients. Am J Clin Oncol. 2003;26(3):285-8.<br />

31. Allo MD, Bennion RS, Kathir K, et al. Ticarcillin/clavulanate versus imipenem/cilistatin for the<br />

treatment of infections associated with gangrenous and perforated appendicitis. Am Surg.<br />

1999;65(2):99-104.<br />

198


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Tetracyclines<br />

AHFS 081224<br />

October 27, 2004<br />

I. Overview<br />

Tetracyclines have been used for the treatment of a wide variety of gram-positive and gramnegative<br />

bacterial infections since the 1950s. They are important for the treatment of and<br />

prophylaxis against infections with bacteria that could be used in biological weapons. Bacterial<br />

resistance to tetracycline was identified shortly after the introduction of therapy. In contrast,<br />

tetracycline resistance has not yet been described in protozoa or other eukaryotic organisms. 10<br />

The tetracyclines were discovered in the 1940s, and because they had few major side effects,<br />

(except in young children and pregnant women) therapeutic use began in the 1950s. At the same<br />

time, tetracyclines were found to be useful growth promoters in animals and have been extensively<br />

used in agriculture in North America since the 1950s. Only a limited number of derivatives are<br />

currently used. Tetracyclines are broad-spectrum agents, with activity against both gram-positive<br />

and gram-negative bacteria and intracellular chlamydiae, mycoplasmas, and rickettsiae. More<br />

recently, tetracyclines have been used against eukaryotic protozoan parasites and for the treatment<br />

of a variety of noninfectious conditions, often at subtherapeutic levels (e.g., for acne) and over<br />

extend time periods. Shortly after the introduction of tetracycline therapy, the first tetracyclineresistant<br />

bacterial pathogen was identified. Since then, tetracycline-resistant bacterial pathogens<br />

have continued to be identified, limiting the tetracyclines’ effectiveness in the treatment of<br />

bacterial disease. 10<br />

That being said, the tetracyclines still play an important role in the treatment of infectious<br />

diseases. These drugs are well absorbed orally and are available generically. They have also<br />

become preferred therapy for some relatively new infectious diseases. Doxycycline is the<br />

preferred agent for rickettsial tick borne diseases and early Lyme disease. 25, 11, 12 Minocycline has<br />

been used to treat MRSA and MRSE infections. 25<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this<br />

review.<br />

Table 1. Tetracyclines in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name (s)<br />

Tetracycline Capsules/Oral suspension *Sumycin, Achromycin V, Emtet-500<br />

Oxytetracycline Parenteral Injection Terramycin<br />

Doxycycline Tablets, Capsules, Injection, Powder for oral<br />

suspension, Syrup<br />

*Vibramycin, Vibra-Tabs, Doryx, Adoxa, ED<br />

Doxy Caps, Monodox, Periostat<br />

Minocycline Tablets, Capsules, Powder for oral suspension,<br />

*Minocin, Dynacin<br />

injection<br />

Demeclocycline Tablets *Declomycin<br />

*Generic Formulation Available.<br />

199


II. Evidence Based Medicine and Current Treatment Guidelines<br />

This category encompasses hundreds of possible treatment algorithms because of the<br />

numerous organisms and infectious diseases that are covered by these drugs. Many<br />

documented treatment protocols and dosage recommendations from a variety of reputable<br />

sources exist. These treatment guidelines can change frequently as resistant strains of<br />

organisms appear. Guidelines are available with the most up-to-date information from the<br />

CDC, 37 Facts and Comparison drug monographs, 9 the various medications prescribing<br />

information for treatment protocols on specific disease states, 1-8 and review articles based on<br />

14, 15,24,27,34,35<br />

accepted practices. Examples of therapy guidelines that include use of<br />

tetracyclines are presented here:<br />

Lyme Disease<br />

For the treatment of early Lyme disease, administration of doxycycline (100mg BID) or<br />

amoxicillin (500mg TID) for 14-21 days is recommended for early localized or early<br />

disseminated Lyme disease associated with erythema migrans, in the absence of neurological<br />

involvement or third-degree atrioventricular heat block (A-I). Doxycycline has the advantage<br />

of being efficacious for the treatment of human granulocytic ehrlichiosis or babesiosis.<br />

Doxycycline is also useful in the treatment of Lyme arthritis.<br />

Sexually Transmitted Diseases<br />

Epididymitis is often caused by C. trachomatis or N. gonorhoeae in sexually active men aged<br />

35 years, men who have recently undergone urinary tract instrumentation or surgery,<br />

and men who have anatomical abnormalities of the urinary tract.<br />

Empiric therapy is indicated before culture results become available. For infections most<br />

likely caused by gonococcal or chlamydial infection, the CDC recommended regimen is:<br />

• Ceftriaxone 250mg IM in a single dose<br />

PLUS<br />

• Doxycycline 100mg orally BID for 10 days<br />

For epididymitis most likely caused by enteric organisms, for patients allergic to<br />

cephalosporins and/or tetracyclines, or for epididymitis in patients aged >35 years, the<br />

recommended regimen is:<br />

• Ofloxacin 300mg orally BID for 10 days<br />

OR<br />

• Levofloxacin 500mg orally QD for 10 days<br />

III. Comparative Indications of the Tetracycline Antibiotics<br />

Table 2. FDA-approved indications for the Tetracycline antibiotics, unlabeled uses, and susceptible<br />

organisms and describes the major organisms under each indication. 9<br />

✔ =Organisms Generally Susceptible to Tetracyclines 1 but not FDA-approved and unlabeled uses.<br />

✔✔=FDA-approved indications<br />

Category or<br />

Disease Organism Demeclocycline Doxycycline Minocycline Oxytetracycline Tetracycline<br />

Gram-positive Actinomyces sp. ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Alpha-hemolytic<br />

streptococci(Viridans<br />

group)<br />

200<br />

✔ ✔ ✔


✔ =Organisms Generally Susceptible to Tetracyclines 1 but not FDA-approved and unlabeled uses.<br />

✔✔=FDA-approved indications<br />

Category or<br />

Disease Organism Demeclocycline Doxycycline Minocycline Oxytetracycline Tetracycline<br />

Bacillus anthracis ✔✔ ✔ ✔✔ ✔✔ ✔✔<br />

Clostridium sp. ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Enterococcus<br />

faecalis 2 , 3 ✔ ✔ ✔ ✔<br />

E. faecium ✔ ✔ ✔<br />

Listeria<br />

monocytogenes<br />

✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Propionibacterium<br />

acnes<br />

✔✔ ✔✔ ✔✔<br />

Staphylococcus<br />

aureus 4 ✔✔ ✔ ✔✔ ✔✔<br />

Streptococcus<br />

pneumoniae 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

S. pyogenes 2 , 5 ✔✔ ✔ ✔ ✔✔ ✔✔<br />

Treponema pallidum ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

T. pertenue ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Gram-negative<br />

Acinetobacter sp. 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Bacteroides sp. 2 ✔✔ ✔ ✔ ✔✔ ✔✔<br />

Bartonella<br />

bacilliformis<br />

✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Borrelia recurrentis ✔✔ ✔✔ ✔✔ ✔✔ ✔<br />

Brucella sp. ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Calymmatobacterium<br />

granulomatis<br />

✔✔ ✔✔ ✔ ✔✔ ✔✔<br />

Campylobacter fetus ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Enterobacter<br />

aerogenes 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔<br />

Escherichia coli 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Francisella tularensis ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Fusobacterium<br />

fusiform<br />

✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Haemophilus ducreyi ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

H. influenzae 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔<br />

Klebsiella sp. 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Neisseria gonorrhoeae ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

N. meningitides ✔ ✔<br />

201


✔ =Organisms Generally Susceptible to Tetracyclines 1 but not FDA-approved and unlabeled uses.<br />

✔✔=FDA-approved indications<br />

Category or<br />

Disease Organism Demeclocycline Doxycycline Minocycline Oxytetracycline Tetracycline<br />

Shigella sp. 2 ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Vibrio cholerae ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Yersinia pestis ✔✔ ✔ ✔✔ ✔✔ ✔✔<br />

Balantidium coli ✔ ✔ ✔<br />

Chlamydia psittaci ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

C. trachomatis ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Entamoeba sp. ✔ ✔ ✔ ✔ ✔<br />

Miscellaneous<br />

Mycobacterium<br />

marinum<br />

Mycoplasma<br />

pneumoniae<br />

Plasmodium<br />

falciparum 6<br />

✔<br />

✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

✔✔<br />

Rickettsiae sp. ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Acute intestinal<br />

amebiasis<br />

Ureaplasma<br />

urealyticum<br />

✔✔ ✔✔ ✔✔<br />

Entamoeba histolytica ✔✔ ✔✔ ✔✔ ✔✔ ✔✔<br />

Severe Acne ✔✔ ✔✔ ✔✔<br />

Anthrax,<br />

including<br />

inhalational<br />

anthrax<br />

Malaria<br />

prophylaxis<br />

Treatment of<br />

asymptomatic<br />

meningococcal<br />

carriers<br />

Periodontitis<br />

Gonococcal<br />

Arthritis, early<br />

Lyme disease,<br />

malaria, ocular<br />

rosacea<br />

Peptic ulcers,<br />

adjunctive<br />

therapy<br />

✔✔<br />

Plasmodium<br />

✔✔<br />

falciparum<br />

N. meningitides ✔✔<br />

Helicobacter pylori<br />

✔<br />

✔<br />

202


✔ =Organisms Generally Susceptible to Tetracyclines 1 but not FDA-approved and unlabeled uses.<br />

✔✔=FDA-approved indications<br />

Category or<br />

Disease Organism Demeclocycline Doxycycline Minocycline Oxytetracycline Tetracycline<br />

Malaria, pleural<br />

malignant<br />

effusions,<br />

nocardiosis,<br />

ocular rosacea,<br />

treatment of<br />

traveler’s<br />

diarrhea,<br />

prophylaxis of<br />

pneumothorax,<br />

✔<br />

Lyme Disease Lyme borreliosis ✔<br />

STD (syphilis,<br />

✔<br />

chlamydia, PID,<br />

epididymitis,<br />

sexual assault<br />

prophylaxis<br />

Rheumatoid<br />

✔<br />

arthritis-early<br />

treatment<br />

Gallbladder E. coli ✔<br />

infections<br />

Nocardiosis,<br />

✔<br />

chronic<br />

malignant pleural<br />

effusion<br />

Syndrome of<br />

✔<br />

inappropriate<br />

antidiuretic<br />

hormone<br />

(SIADH)<br />

Peptic ulcers H. Pylori ✔ ✔✔<br />

1 Cross-resistance of these organisms to tetracyclines is common.<br />

2 Because many strains of gram-negative micro-organisms have been shown to be resistant to tetracyclines, culture and susceptibility<br />

testing are recommended.<br />

3 Up to 74% of Enterococcus faecalis have been found to be resistant to tetracyclines.<br />

4 Tetracyclines are not the drugs of choice in the treatment of any type of staphylococcal infections.<br />

5 Up to 44% of Streptococcus pyogenes have been found to be resistant to tetracycline drugs.<br />

6 Doxycycline has been found to be active against the asexual erythrocytic form of Plasmodium falciparum but not against the<br />

gametocytes of P. falciparum.<br />

IV. Pharmacokinetic Parameters 1-9<br />

Tetracyclines are adequately, but incompletely, absorbed from the GI tract. The percentage<br />

absorbed when taken on an empty stomach is lowest for oxytetracycline, demeclocycline, and<br />

tetracycline, and highest for doxycycline and minocycline. The extent of absorption is usually<br />

decreased by the presence of divalent and trivalent cations, and to a variable degree by milk or<br />

food (see Drug Interactions). Tetracyclines are bound to plasma proteins in varying degrees.<br />

203


Penetration of the tetracyclines into most body fluids and tissues is excellent. Tetracyclines are<br />

distributed in varying amounts into bile, liver, lung, kidney, prostate, urine, CSF, synovial fluid,<br />

mucosa of the maxillary sinus, brain, sputum, and bone. Inflammation of the meninges is not<br />

required for passage into the CSF, but concentrations may increase in the presence of inflamed<br />

meninges. Tetracyclines cross the placenta and enter the fetal circulation and amniotic fluid.<br />

The tetracyclines are concentrated in the bile by the liver. They are excreted in the urine and feces<br />

at high concentrations in a biologically active form. Because renal clearance of tetracyclines is by<br />

glomerular filtration, excretion is significantly affected by the state of renal function. The renal<br />

clearance of demeclocycline has been shown to be about half of that of tetracycline. The urinary<br />

and fecal recovery of minocycline is one-half to one-third that of other tetracyclines, and<br />

minocycline also appears to undergo some metabolism, largely to 9-hydroxyminocycline.<br />

Doxycycline appears to be excreted extensively by the digestive tract.<br />

Table 4. Pharmacokinetic Parameters of the Tetracycline Agents 1-9<br />

* nd = no data.<br />

1 300 mg single oral dose.<br />

2 200 mg single oral dose.<br />

3 200 mg administered IV over 2 hours.<br />

4 Single oral dose of two 100 mg pellet-filled capsules.<br />

Protein<br />

binding<br />

(%)<br />

Serum<br />

half-life<br />

(h)<br />

Excreted<br />

in<br />

urine (%)<br />

Tetracyclines<br />

Absorption<br />

(%) C max (mcg/mL) T max (h)<br />

Demeclocycline 60 to 80 1.5 to 1.7 1 3 to 4 1 35 to 90 16 nd *<br />

Doxycycline 90 to 100 2.6 (hyclate) 2 2 (hyclate) 2 80 to 95 to 22 40<br />

3.6 (IV) 3<br />

3.61(monohydrate) 2 2.6(monohydrate) 2<br />

Minocycline 90 to 100 2.1 to 5.1 4 1 to 4 75 11 to 22 5 to 10<br />

(oral)<br />

15 to 23<br />

(IV)<br />

Oxytetracycline 60 to 80 nd 2 to 4 20 to 40 6 to 12 10 to 35<br />

Tetracycline 60 to 80 nd 2 to 4 20 to 65 6 to 12 20 to 55<br />

204


V. Drug Interactions<br />

Table 5. Drug Interactions of the Tetracycline Agents 1- 9<br />

Precipitant drug Object drug * Description<br />

Antacids<br />

(containing<br />

aluminum, calcium<br />

or magnesium<br />

salts)<br />

Iron salts<br />

Zinc salts<br />

Tetracyclines<br />

Tetracyclines administered with aluminum, calcium, magnesium,<br />

iron, or zinc salts form an insoluble chelate, thereby decreasing<br />

the absorption and serum levels of the tetracycline. Administer<br />

tetracyclines at least 2 hours before or after these agents.<br />

Barbiturates Doxycycline Barbiturates increase the hepatic metabolism of doxycycline,<br />

therefore decreasing doxycycline’s half-life and serum levels.<br />

Adjust doxycycline dose as needed. Consider using an<br />

alternative tetracycline.<br />

Bismuth salts Tetracyclines Coadministration of bismuth salts in liquid formulations may<br />

decrease the serum levels of tetracyclines. Give the bismuth salt<br />

2 hours after the tetracycline.<br />

Carbamazepine Doxycycline Carbamazepine may decrease the half-life and serum levels of<br />

doxycycline due to increased hepatic metabolism. Adjust<br />

doxycycline dose as needed. Consider using an alternative<br />

tetracycline.<br />

Cholestyramine<br />

Colestipol<br />

Phenytoin<br />

Rifamycins<br />

Urinary alkalinizers<br />

(e.g., sodium<br />

lactate, potassium<br />

citrate)<br />

Tetracyclines<br />

Tetracyclines<br />

Tetracyclines<br />

Doxycycline<br />

Tetracyclines<br />

Anticoagulants,<br />

oral<br />

Contraceptives,<br />

oral<br />

Coadministration may decrease or delay the absorption of<br />

tetracyclines, therefore decreasing the serum concentrations.<br />

Adjust the tetracycline dose if needed.<br />

Phenytoin and rifamycins appear to induce the metabolism of<br />

doxycycline, causing the half-life to be significantly decreased.<br />

Increased doxycycline dosage may be needed.<br />

Coadministration may result in increased excretion of the<br />

tetracyclines and decreased serum levels. Separate<br />

administration by 3 to 4 hours; however, this may not be<br />

effective and an increase in tetracycline dose may be necessary if<br />

the pH of the urine remains increased.<br />

The action of oral anticoagulants may be increased because of<br />

the elimination of vitamin K-producing gut bacteria by<br />

tetracyclines. Monitor coagulation parameters and adjust<br />

anticoagulant dose as needed.<br />

Tetracyclines may interfere with the enterohepatic recirculation<br />

of certain contraceptive steroids, leading to reduced efficacy.<br />

Although infrequently reported, contraceptive failure is possible.<br />

Tetracyclines Digoxin Coadministration may result in increased serum levels of digoxin<br />

in a small subset of patients ( 10%). Monitor digoxin levels and<br />

signs of toxicity.<br />

Tetracyclines Insulin The ability of insulin to produce hypoglycemia may be<br />

potentiated. In diabetic patients, monitor blood glucose<br />

concentrations closely and tailor the insulin regimen as needed.<br />

Tetracyclines Isotretinoin Isotretinoin use has been associated with a number of cases of<br />

pseudotumor cerebri, some of which involved coadministration<br />

of tetracyclines. Therefore, avoid concomitant use.<br />

Tetracyclines Methoxyflurane Coadministration may enhance the risk for renal toxicity; deaths<br />

have been reported. Do not coadminister. If possible seek<br />

alternative agents.<br />

Tetracyclines Penicillins The bacteriostatic action of tetracyclines may interfere with the<br />

205


Precipitant drug Object drug * Description<br />

bactericidal activity of penicillins. Consider avoiding this<br />

combination if at all possible.<br />

Tetracyclines Theophyllines The incidence of adverse reactions to theophyllines may be<br />

increased. Monitor theophylline levels and adjust dose as<br />

needed.<br />

*<br />

= Object drug increased. = Object drug decreased.<br />

Drug / Food Interactions: 1-9<br />

The administration of demeclocycline, oxytetracycline, and tetracycline with milk and dairy<br />

products forms poorly absorbed chelates. A number of studies have reported the serum levels of<br />

these tetracyclines, when administered with milk products, to be 50% to 80% lower. Administer<br />

the interacting tetracyclines at least 2 hours before or after meals. The inhibitory effect of food<br />

and milk on the absorption of doxycycline and minocycline is considerably less than that observed<br />

with the other tetracycline derivatives. These two drugs are often administered without regard to<br />

meals; but the potential risk of decreased drug efficacy must be weighed against the benefit of<br />

treating the infection. The administration of doxycycline with a high-fat meal has been shown to<br />

delay the time to peak plasma concentrations by an average 1 hour 20 minutes. Peak plasma<br />

concentrations of doxycycline were also decreased by up to 20% with simultaneous ingestion of<br />

dairy products or a high-fat, high-protein meal. The peak plasma concentration of minocycline<br />

was slightly decreased and delayed by 1 hour when administered with food, compared to dosing<br />

under fasting conditions.<br />

VI. Adverse Drug Events of the Tetracycline Agents<br />

ADRs are possible with all agents in the class unless specified.<br />

Table 6. Common Adverse Events Reported for the Tetracycline Agents 1-9<br />

Category<br />

Adverse Event<br />

ORAL<br />

CNS<br />

Dermatologic<br />

GI<br />

Dizziness, headache, bulging fontanel, pseudotumor cerebri, convulsions, hypesthesia,<br />

paresthesia, sedation, vertigo, myasthenic syndrome (demeclocycline; rare).<br />

Maculopapular and erythematous rashes, photosensitivity, fixed drug eruptions, balanitis,<br />

erythema multiforme, Stevens-Johnson syndrome, skin and mucus membrane pigmentation,<br />

alopecia, erythema nodosum, hyperpigmentation of the nails, pruritus, toxic epidermal<br />

necrolysis, vasculitis; exfoliative dermatitis (rare).<br />

Anorexia, nausea, vomiting, diarrhea, glossitis, dysphagia, enterocolitis, inflammatory lesions<br />

(with monilial overgrowth) in the anogenital region, esophageal ulcerations, pancreatitis,<br />

dyspepsia, stomatitis, enamel hypoplasia, pseudomembranous colitis, esophagitis, bulky loose<br />

stools, sore throat, black hairy tongue, hoarseness (tetracycline).<br />

Due to oral minocycline and doxycycline’ s virtually complete absorption, side effects of the<br />

lower bowel, particularly diarrhea, have been infrequent.<br />

Hematologic<br />

Hepatic<br />

Hypersensitivity<br />

Anemia, hemolytic anemia, thrombocytopenia, neutropenia, eosinophilia<br />

Increased liver enzymes, hepatic toxicity, hyperbilirubinemia, hepatic cholestasis; hepatic<br />

failure, hepatitis (rare).<br />

Urticaria, angioneurotic edema, pericarditis, anaphylaxis, anaphylactoid purpura, systemic lupus<br />

erythematous exacerbation, polyarthralgia, pulmonary infiltrates with eosinophilia.<br />

Musculoskeletal<br />

Arthralgia, arthritis, bone discoloration, myalgia, joint stiffness and swelling.<br />

206


Renal<br />

Respiratory<br />

Category<br />

Miscellaneous<br />

PARENTERAL<br />

CNS<br />

Dermatologic<br />

GI<br />

Hematologic<br />

Hepatic<br />

Hypersensitivity<br />

Musculoskeletal<br />

Renal<br />

Respiratory<br />

Miscellaneous<br />

Adverse Event<br />

Dose-related increase in BUN, acute renal failure, interstitial nephritis, nephrogenic diabetes<br />

insipidus (demeclocycline).<br />

Cough, dyspnea, bronchospasm, asthma exacerbation.<br />

Brown-black microscopic discoloration of thyroid glands (prolonged therapy), tooth<br />

discoloration, lupus-like syndrome, fever, secretion discoloration, vulvovaginitis, tinnitus,<br />

decreased hearing, serum sickness-like syndrome.<br />

Headache, convulsions, dizziness, hypesthesia, paresthesia, sedation, vertigo, bulging fontanels,<br />

pseudotumor cerebri<br />

Maculopapular and erythematous rashes, photosensitivity, alopecia, erythema nodosum,<br />

hyperpigmentation of the nails, pruritus, toxic epidermal necrolysis, vasculitis, fixed drug<br />

eruptions, balanitis, erythema multiforme, Stevens-Johnson syndrome, skin and mucus<br />

membrane pigmentation, injection site erythema and injection site pain, exfoliative dermatitis<br />

(rare).<br />

Anorexia, nausea, vomiting, diarrhea, glossitis, dysphagia, enterocolitis, inflammatory lesions<br />

(with monilial overgrowth) in the anogenital region, dyspepsia, stomatitis, enamel hypoplasia,<br />

pseudomembranous colitis, pancreatitis.<br />

Hemolytic anemia, thrombocytopenia, neutropenia, eosinophilia, agranulocytosis, leukopenia,<br />

pancytopenia.<br />

Hyperbilirubinemia, hepatic cholestasis, increased liver enzymes, jaundice, hepatitis, liver failure<br />

Urticaria, angioneurotic edema, anaphylaxis, anaphylactoid purpura, pericarditis, exacerbation of<br />

systemic lupus erythematous, myocarditis, pulmonary infiltrates.<br />

Arthralgia, arthritis, bone discoloration, myalgia, joint stiffness and swelling, polyarthralgia<br />

Dose-related increase in BUN; interstitial nephritis, acute renal failure(minocycline).<br />

Cough, dyspnea, bronchospasm, asthma exacerbation<br />

Brown-black microscopic discoloration of thyroid glands (prolonged therapy);tooth<br />

discoloration, vulvovaginitis, tinnitus, hypersensitivity syndrome (cutaneous reaction,<br />

eosinophilia, and one or more of the following: Hepatitis, pneumonitis, nephritis, myocarditis,<br />

pericarditis, fever, lymphadenopathy), lupus-like syndrome, serum sickness-like syndrome,<br />

fever, secretion discoloration<br />

Warnings 1-9<br />

Malaria prophylaxis (doxycycline only):<br />

Doxycycline offers substantial but not complete suppression of the asexual stages of Plasmodium<br />

strains. It does not suppress P. falciparum's sexual blood stage gametocytes and therefore patients<br />

completing this prophylactic regimen may still transmit the infection to mosquitos outside<br />

endemic areas. Advise patients taking doxycycline for malaria prophylaxis of when prophylaxis<br />

should begin and end; that no present-day antimalarial, including doxycycline, guarantees<br />

protection against malaria; and to avoid being bitten by mosquitos by wearing protective clothing,<br />

using effective insect-repellent, mosquito nets, etc.<br />

Pseudomembranous colitis:<br />

Treatment with antibacterial agents alters the normal flora of the colon and may permit<br />

overgrowth of clostridia. Pseudomembranous colitis has been reported with nearly all<br />

antibacterial agents and may range in severity from mild to life-threatening. It is important to<br />

consider this diagnosis in patients who present with diarrhea following subsequent administration<br />

of antibacterial agents. Once the diagnosis is established, initiate therapeutic measures. Mild cases<br />

usually respond to discontinuation of the drug. Moderate to severe cases may require management<br />

with fluids, electrolytes, protein supplementation, and treatment with an antibacterial agent<br />

effective against Clostridium difficile colitis.<br />

207


Parenteral therapy:<br />

Reserve for situations in which oral therapy is not indicated. Institute oral therapy as soon as<br />

possible. If given IV over prolonged periods, thrombophlebitis may result. IM use produces<br />

lower blood levels than recommended oral dosages. If high blood levels are needed rapidly,<br />

administer IV.<br />

Nephrogenic diabetes insipidus:<br />

Administration of demeclocycline has resulted in appearance of the diabetes insipidus syndrome<br />

(e.g., polyuria, polydipsia, weakness) in some patients on long-term therapy. The syndrome has<br />

been shown to be nephrogenic, dose-dependent, and reversible upon discontinuation of therapy.<br />

Hypersensitivity reactions:<br />

Sensitivity reactions are more likely to occur on patients with a history of allergy, asthma, hay<br />

fever, or urticaria. Use tetracyclines with caution in these patients. Cross-sensitivity among the<br />

tetracyclines is extremely common.<br />

Renal function impairment:<br />

Use tetracyclines with caution in patients with impaired renal function.<br />

If renal impairment exists, even usual doses may lead to excessive systemic accumulation of the<br />

tetracyclines (with the exception of doxycycline) and possible liver toxicity. Use lower than usual<br />

doses; if therapy is prolonged, drug serum level determinations may be advisable. Concurrent use<br />

of tetracycline and methoxyflurane has resulted in fatal renal toxicity.<br />

The antianabolic action of tetracyclines may cause an increase in blood urea nitrogen. In<br />

significantly impaired renal function, higher serum tetracycline levels may lead to azotemia,<br />

hyperphosphatemia, and acidosis. This does not seem to occur with doxycycline.<br />

Hepatic function impairment:<br />

Use tetracyclines with caution in patients with impaired liver function.<br />

In the presence of renal dysfunction, and particularly in pregnancy, IV tetracycline more than 2<br />

g/day has been associated with death secondary to liver failure. When need for intensive treatment<br />

outweighs its potential dangers (especially during pregnancy or in known or suspected renal and<br />

liver impairment), monitor renal and liver function tests. Serum tetracycline concentrations should<br />

not exceed 15 mcg/mL. Do not prescribe other potentially hepatotoxic drugs concomitantly.<br />

Hepatotoxicity has been reported with minocycline; therefore, minocycline should be used with<br />

caution in patients with hepatic dysfunction and in conjunction with other hepatotoxic drugs.<br />

The hazard of liver toxicity is of particular importance in parenteral administration to pregnant or<br />

postpartum patients with pyelonephritis.<br />

Carcinogenesis:<br />

There has been evidence of oncogenic activity in studies with oxytetracycline (adrenal and<br />

pituitary tumors) in rats and minocycline (thyroid tumors) in rats and dogs.<br />

Mutagenesis:<br />

Tetracycline and oxytetracycline have produced positive mutagenic results in mammalian cell<br />

assays in vitro.<br />

Fertility impairment:<br />

Minocycline has been shown to impair fertility in male rats.<br />

Pregnancy:<br />

Category D. Tetracyclines readily cross the placenta and are found in fetal tissues and can have<br />

toxic effects on the developing fetus (retardation of skeletal development). Evidence of<br />

embryotoxicity has also been noted in animals treated early in pregnancy.<br />

208


A case-control study (18,515 mothers of infants with congenital anomalies and 32,804 mothers of<br />

infants with no congenital anomalies) shows a weak but marginally statistically significant<br />

association with total malformations and use of doxycycline any time during pregnancy. Sixtythree<br />

(0.19%) of the controls and 56 (0.3%) of the cases were treated with doxycycline. This<br />

association was not seen when the analysis was confined to maternal treatment during the period<br />

of organogenesis with the exception of a marginal relationship with neural tube defect based on<br />

only two exposed cases.<br />

Lactation:<br />

Tetracyclines are excreted in breast milk. Milk:plasma ratios vary between 0.25 and 1.5. Because<br />

of the potential for serious adverse reactions, decide whether to discontinue nursing or discontinue<br />

the drug.<br />

Children:<br />

Generally, do not use tetracyclines in children under eight years of age (except for anthrax,<br />

including inhalational) due to adverse effects on teeth and bones, unless other drugs are not likely<br />

to be effective or are contraindicated.<br />

Teeth:<br />

The use of tetracyclines during the period of tooth development (from the last half of pregnancy<br />

through 8 years of age) may cause permanent discoloration (yellow, gray, brown) of teeth. This<br />

adverse reaction is more common during long-term use of the drugs, but has been observed<br />

following repeated short-term courses. Enamel hypoplasia has also been reported.<br />

Bone:<br />

Tetracyclines form a stable calcium complex in any bone-forming tissue. Decreased fibula growth<br />

rate occurred in premature infants given 25 mg/kg oral tetracycline every 6 hours. This was<br />

reversible when the drug was discontinued.<br />

Photosensitivity:<br />

Photosensitivity manifested by an exaggerated sunburn reaction has been observed in some<br />

individuals taking tetracyclines. Advise patients who are apt to be exposed to direct sunlight or<br />

ultraviolet light that this reaction can occur with tetracycline drugs, and discontinue treatment at<br />

the first evidence of skin erythema.<br />

Exaggerated sunburn reactions are characterized by severe burns of exposed surfaces, resulting<br />

from direct exposure to sunlight during therapy with moderate or large doses. Phototoxic<br />

reactions are most frequent with demeclocycline, and occur less frequently with the other<br />

tetracyclines.<br />

209


VII.<br />

Dosing and Administration for the Tetracycline Agents<br />

Table 7. Dosing for the Tetracycline Agents 5-7<br />

Drug Availability Dose /Frequency/Duration<br />

Tetracycline<br />

Capsules: 250mg<br />

Capsules: 500mg<br />

Oral Suspension:125mg/5mL<br />

Take with plenty of fluids. Food and some dairy products interfere with the absorption of<br />

tetracycline.<br />

Adults:<br />

Usual dose: 1 to 2g/day in 2 or 4 equal doses.<br />

Oxytetracycline<br />

Doxycycline<br />

Minocycline<br />

Injection:50mg/mL with 2% lidocaine<br />

125mg/mL with 2% lidocaine<br />

Lidocaine is included in this formulation not as a therapeutic agent, but<br />

to decrease the pain associated with intramuscular injection.<br />

Tablets: 100mg (as hyclate)<br />

Capsules: 50mg (as hyclate)<br />

Capsules: 100mg (as hyclate)<br />

Capsules, coated pellets: 75mg (as hyclate)<br />

100mg (as hyclate)<br />

Tablets: 50mg (as monohydrate)<br />

Tablets: 75mg (as monohydrate)<br />

100mg (as monohydrate)<br />

Capsules: 50mg (as monohydrate)<br />

100mg (as monohydrate)<br />

Powder for Oral Suspension: 25mg (as monohydrate) per<br />

5mL when reconstituted<br />

Syrup: 50mg (as calcium) per 5mL<br />

Powder for Injection, lyophilized: 100mg (as hyclate)<br />

Powder for Injection, lyophilized: 200mg (as hyclate)<br />

Tablets: 50mg (as HCl)<br />

75mg (as HCl)<br />

100mg (as HCl)<br />

Capsules: 50mg (as HCl)<br />

Capsules: 75mg (as HCl)<br />

See package insert for dosing on specific diseases.<br />

The usual daily dose is 250mg administered IM once every 24 hours or 300mg given in divided<br />

doses at 8 to 12 hour intervals.<br />

Oral:<br />

When used in streptococcal infections, continue therapy for 10 days. Take with plenty of<br />

water. Absorption and peak plasma levels may be reduced when administered with meals<br />

or with dairy products, including milk.<br />

Adults:<br />

Usual dose:<br />

200 mg on the first day of treatment (100mg every 12 hours); follow with a maintenance<br />

dose of 100mg/day. The maintenance dose may be administered as a single dose or as 50<br />

mg every 12 hours.<br />

More severe infections (particularly chronic urinary tract infections):<br />

100mg every 12 hours.<br />

See package insert for dosing on specific diseases.<br />

Oral:<br />

May be taken with or without food. Take with plenty of fluids.<br />

Adults:<br />

Usual dosage:


Drug Availability Dose /Frequency/Duration<br />

Capsules: 100mg (as HCl)<br />

Capsules, pellet filled: 50mg (as HCl)<br />

100mg (as HCl)<br />

Oral suspension:50mg (as HCl)/5 mL<br />

Powder for injection, cryodesiccated: 100mg<br />

200mg initially, followed by 100mg every 12 hours. If more frequent doses are preferred, give 100<br />

or 200mg initially; follow with 50mg, 4 times/day.<br />

See package insert for dosing on specific diseases.<br />

Demeclocycline 150mg and 300mg tablets Take with plenty of fluids. Foods and some dairy products interfere with absorption; take<br />

demeclocycline at least 1 hour before or 2 hours after meals or dairy products.<br />

Adults:<br />

Daily dose:<br />

4 divided doses of 150mg each or 2 divided doses of 300mg each.<br />

Gonorrhea patients sensitive to penicillin:<br />

Initially, 600mg; follow with 300mg every 12 hours for 4 days to a total of 3 g.<br />

Streptococcal infections:<br />

Treat streptococcal infections for at least 10 days.<br />

Concomitant therapy:<br />

Absorption is impaired by antacids containing aluminum, calcium, or magnesium, and by<br />

preparations containing iron. Take demeclocycline at least 1 hour before or 2 hours after<br />

these products.<br />

211


Special Dosing Considerations<br />

Table 8. Special Dosing Considerations for the Tetracycline Agents 1- 9<br />

Drug Renal Dosing Hepatic Dosing Pediatric Use Pregnancy<br />

Category<br />

Tetracycline<br />

Decrease recommended<br />

dosages and/or extend dosing<br />

intervals in patients with renal<br />

impairment.<br />

Children (over 8 years of age):<br />

Daily dose is 10 to 20mg/lb (25 to<br />

50mg/kg) in 4 equally divided doses.<br />

D<br />

Can Drug Be<br />

Crushed<br />

Yes<br />

Oxytetracycline<br />

Renal function impairment:<br />

Decrease recommended dosage<br />

and/or extend dosing intervals<br />

in patients with renal<br />

impairment.<br />

Children (over 8 years of age):<br />

15 to 25mg/kg, up to a maximum of<br />

250mg per single daily IM injection.<br />

Dosage may be divided and given at 8<br />

to 12 hour intervals.<br />

D<br />

N/A<br />

Doxycycline Children (over 8 years of age):<br />

100lb or less (less than 45kg):<br />

2mg/lb (4.4 mg/kg) divided into<br />

2 doses on the first day of treatment;<br />

follow with 1mg/lb (2.2mg/kg) given<br />

as a single daily dose or divided into 2<br />

doses on subsequent days.<br />

More severe infections:<br />

Up to 2mg/lb (4.4mg/kg) may be used.<br />

For children over 100lb (45kg):<br />

Use the usual adult dose.<br />

D<br />

Yes<br />

Minocycline<br />

Decrease the recommended dosage<br />

and/or increase the dosing<br />

intervals in patients with renal<br />

impairment. Do not exceed<br />

200mg Minocin in 24 hours in<br />

patients with renal impairment<br />

Children (over 8 years of age):<br />

Initially, 4mg/kg; follow with 2mg/kg<br />

every 12 hours.<br />

D<br />

Yes<br />

212


Drug Renal Dosing Hepatic Dosing Pediatric Use Pregnancy<br />

Category<br />

Demeclocycline Renal/Hepatic function<br />

Renal/Hepatic function Children (over 8 years of age):<br />

D<br />

impairment:<br />

impairment:<br />

Usual daily dose:<br />

Administer tetracyclines<br />

Administer tetracyclines<br />

3 to 6mg/lb (6.6 to 13.2mg/kg),<br />

cautiously with renal or hepatic cautiously with renal or<br />

depending upon the severity of<br />

impairment; reduce the<br />

hepatic impairment; reduce the disease, divided into 2 or 4<br />

recommended dosage and/or the recommended dosage<br />

doses.<br />

extend the dosing interval. and/or extend the dosing<br />

interval.<br />

Can Drug Be<br />

Crushed<br />

Yes<br />

213


VIII.<br />

Comparative Effectiveness of the Tetracycline Agents*<br />

Table 9. Additional Outcomes Evidence for the Tetracyclines<br />

Study Sample Treatment /<br />

Duration<br />

Wormser n=180 Ten days of<br />

al. 12<br />

GP, et<br />

oral<br />

doxycycline,<br />

with or without<br />

a single<br />

intravenous<br />

dose of<br />

ceftriaxone, or<br />

20 days of oral<br />

doxycycline<br />

Results<br />

A randomized, double-blind, placebo-controlled trial of patients with early<br />

Lyme Disease.<br />

• At all time points, the complete response rate was similar for<br />

the three treatment groups in both on-study and intention-totreat<br />

analyses.<br />

• There were no significant differences in the results of<br />

neurocognitive testing among the three treatment groups and a<br />

separate control group without Lyme disease.<br />

• Diarrhea occurred significantly more often in the doxycyclineceftriaxone<br />

group (35%) than in either of the other two groups.<br />

Tilley<br />

BC, et<br />

al. 13 n=219 A double-blind,<br />

randomized,<br />

multicenter, 48-<br />

week trial of<br />

oral<br />

minocycline<br />

(200mg/d) or<br />

placebo.<br />

CONCLUSION: Extending treatment with doxycycline from 10 to 20 days<br />

or adding one dose of ceftriaxone to the beginning of a 10-day course of<br />

doxycycline did not enhance therapeutic efficacy in patients with erythema<br />

migrans. Regardless of regimen, objective evidence of treatment failure was<br />

extremely rare.<br />

219 patients with active rheumatoid arthritis participated in a 48 week,<br />

double-blind, placebo-controlled trial (MIRA trial).<br />

• 109 and 110 patients were randomly assigned to receive<br />

minocycline and placebo, respectively. At entry, demographic,<br />

clinical, and laboratory measurements were similar in both<br />

groups. Most patients had mild to moderate disease activity and<br />

some evidence of destructive disease.<br />

• At the week 48 visit, 79% of the minocycline group and 78% of<br />

the placebo group continued to receive the study medication.<br />

At 48 weeks, more patients in the minocycline group than in the<br />

placebo group showed improvement in joint swelling (54% and<br />

39%) and joint tenderness (56% and 41%) (p < 0.023 for both<br />

comparisons). The minocycline group also showed greater<br />

improvement in hematocrit, erythrocyte sedimentation rate,<br />

platelet count, and IgM rheumatoid factor levels (all P values <<br />

0.001), and more patients receiving minocycline had laboratory<br />

values within normal ranges at 48 weeks.<br />

• For the remaining outcomes, P values ranged from 0.04 to 0.76,<br />

all greater than the critical value of 0.005 (Bonferroni<br />

adjustment for multiple comparisons). The frequency of<br />

reported side effects was similar in both groups, and no serious<br />

toxicity occurred.<br />

CONCLUSION: Minocycline was safe and effective for patients with mild<br />

to moderate rheumatoid arthritis. Its mechanisms of action remain to be<br />

determined.<br />

214


Study Sample Treatment /<br />

Results<br />

Duration<br />

Stone M,<br />

et al. 26<br />

535-metaanalysis<br />

1966-2002 The purpose of this analysis was to compare the effectiveness of tetracycline<br />

antibiotics versus control (placebo or conventional treatment) in rheumatoid<br />

arthritis.<br />

• Tetracyclines, when administered for > 3 months, were associated<br />

with a significant reduction in disease activity in RA as follows: for<br />

TJC, standardized mean difference (SMD) = -0.39, 95% CI -0.74, -<br />

0.05; and for acute phase reactants, ESR, SMD = -8.96, 95% CI -<br />

14.51, -3.42. The treatment effect was more marked in the subgroup<br />

of patients with disease duration < 1 year who were seropositive.<br />

• There was no absolute increased risk of adverse events associated<br />

with tetracyclines: absolute risk difference = 0.10, 95% confidence<br />

interval (CI) -0.01, 0.21. No beneficial effect was seen on<br />

radiological progression of disease: for erosions, SMD = 0.17, 95%<br />

CI -0.29, 0.64.<br />

• In addition, subgroup analysis excluding trials with doxycycline<br />

showed that minocycline alone had a greater effect on reduction of<br />

disease activity: for TJC, SMD = -0.69, 95% CI -0.89, -0.49; and<br />

for ESR, SMD = -10.14, 95% CI -14.72, -5.57.<br />

Maesen<br />

FP, et<br />

al. 28 n=41 Doxycycline or<br />

minocycline<br />

100mg BID for<br />

seven days<br />

Kovacs N=103 Minocycline or<br />

al. 30 10 days<br />

GT, et<br />

doxycycline for<br />

CONCLUSION: Tetracyclines, in particular minocycline, were associated<br />

with a clinically significant improvement in disease activity in RA with no<br />

absolute increased risk of side effects. Unfortunately, the information<br />

available was inadequate to allow a detailed analysis of individual side<br />

effects in the studies. Further research is warranted to compare these agents<br />

to newer disease modifying drugs for comparable safety, efficacy, and costeffectiveness.<br />

This double-blind comparative clinical trial observed patients admitted to the<br />

hospital because of acute purulent exacerbations of chronic respiratory<br />

disease.<br />

• Bacteriological and clinical assessment before and immediately after<br />

treatment showed no significant differences between the<br />

doxycycline and the minocycline groups, nor did further evaluation<br />

after seven days follow-up.<br />

• Pharmacokinetic studies showed that the Cmax and 0-11 h AUC<br />

values in blood were higher for doxycycline, whereas the sputum<br />

Cmax was, on average, higher for minocycline because of the<br />

greater penetration of the latter.<br />

• The MIC values for the two antibiotics differed slightly, usually, but<br />

not always, in favor of minocycline.<br />

• Problems were experienced with both agents in the eradication of<br />

Haemophilus influenzae.<br />

CONCLUSION: The net clinical results with the two drugs were identical.<br />

One hundred and three women were found to have Chl. trachomatis infection<br />

of the cervix and were invited to participate in a clinical trial of minocycline<br />

and doxycycline for the treatment of chlamydial infection.<br />

• A 10-day course of either drug resulted in a negative result of a<br />

chlamydial culture for all patients at the follow-up assessment,<br />

which occurred between 11 days to 12 weeks after the therapy.<br />

• Minocycline appeared to have a slight advantage with respect to the<br />

resolution of the gynecological symptoms that were associated with<br />

the chlamydial infection.<br />

215


Study Sample Treatment /<br />

Duration<br />

Ramano<br />

wski b, et<br />

al. 31 N=253 Minocycline<br />

100mg nightly<br />

or doxycycline<br />

100mg BID for<br />

7 days<br />

Results<br />

CONCLUSION: Minocycline and doxycycline showed equal effectiveness<br />

in the eradication of mycoplasmas in over 80% of the treated patients.<br />

The objective in this randomized, double-blind trial was to compare the<br />

efficacy and tolerability of minocycline versus doxycycline in the treatment<br />

of nongonococcal urethritis and mucopurulent cervicitis.<br />

• 151 men and 102 women with nongonococcal urethritis,<br />

mucopurulent cervicitis or whose sexual partner had either condition<br />

or a positive culture for Chlamydia trachomatis.<br />

• The proportion with urethritis or cervicitis did not differ by<br />

treatment group at any follow-up visit (P > 0.08). Unprotected<br />

sexual contact did not affect clinical or microbiological cure rates.<br />

• Adverse effects occurred more frequently in the doxycycline group<br />

(men: 43% versus 26%; P = 0.05; women: 62% versus 35%; P =<br />

0.009). Although the proportion with dizziness did not differ by<br />

drug administered (P = 0.1), dizziness was reported more often by<br />

women (11% versus 3%).<br />

Shapiro Various Retrospective<br />

L, et al. 19 literature<br />

review<br />

Langevit<br />

z P, et<br />

al. 20 <strong>Review</strong> Minocycline<br />

given for<br />

various times<br />

and dosages in<br />

three double-<br />

CONCLUSION: Minocycline, 100mg nightly, was as effective as<br />

doxycycline, 100mg twice daily, each given for seven days in the treatment<br />

of nongonococcal urethritis and mucopurulent cervicitis. Vomiting and<br />

gastrointestinal upset occurred more frequently in the doxycycline group.<br />

Background: Because minocycline can cause serious adverse events,<br />

including hypersensitivity syndrome reaction (HSR), serum sickness-like<br />

reaction (SSLR), and drug-induced lupus, a follow-up study based on a<br />

retrospective review of our Drug Safety Clinic and the Health Protection<br />

Branch databases and a literature review was conducted to determine if<br />

similar rare events are associated with tetracycline and doxycycline. Cases<br />

of isolated single organ dysfunction (SOD) attributable to the use of these<br />

antibiotics also were identified.<br />

• Nineteen cases of HSR due to minocycline, two due to tetracycline,<br />

and one due to doxycycline were identified. Eleven cases of SSLR<br />

due to minocycline, three due to tetracycline, and two due to<br />

doxycycline were identified. All 33 cases of drug-induced lupus<br />

were attributable to minocycline. Forty cases of SOD from<br />

minocycline, 37 cases from tetracycline, and six from doxycycline<br />

were detected. Hypersensitivity syndrome reaction, SSLR, and<br />

SOD occur on average within four weeks of therapy, whereas<br />

minocycline-induced lupus occurs on average two years after the<br />

initiation of therapy.<br />

CONCLUSION: Early serious events occurring during the course of<br />

tetracycline antibiotic treatment include HSR, SSLR, and SOD. Druginduced<br />

lupus, which occurs late in the course of therapy, is reported only<br />

with minocycline. We theorize that minocycline metabolism may account<br />

for the increased frequency of serious adverse events with this drug.<br />

<strong>Review</strong> of three double-blind studies giving minocycline to patients with<br />

rheumatoid arthritis.<br />

• The antirheumatic effect of minocycline can be related to its<br />

immunomodulatory and anti-inflammatory, rather than to its<br />

antibacterial properties. Its efficacy in rheumatoid arthritis has been<br />

216


Study Sample Treatment /<br />

Duration<br />

blind studies<br />

Results<br />

reported in two open trials and in three double-blind controlled<br />

studies.<br />

• The first two double-blind studies, one in The Netherlands and one<br />

in the US, were performed in patients with advanced disease. Both<br />

studies showed a modest, but statistically significant improvement<br />

in the clinical parameters of disease activity and in the erythrocyte<br />

sedimentation rate in the minocycline-treated patients.<br />

• The US study also reported that patients in the minocycline group<br />

developed fewer types of erosion than those in the placebo group.<br />

This finding supports the role of minocycline as a disease modifying<br />

agent.<br />

• The common adverse effects of minocycline reported in these two<br />

studies included gastrointestinal adverse effects, dizziness, rash, and<br />

headaches. Less common adverse effects were intracranial<br />

hypertension, pneumonitis, persistent skin and mucosal<br />

hyperpigmentation, lupus-like syndrome and acute hepatic injury.<br />

• The third double-blind study enrolled only seropositive rheumatoid<br />

arthritis patients with early disease (less than one year duration), and<br />

showed very encouraging results of significant improvement in the<br />

disease activity parameters in the minocycline treated group of<br />

patients. The same authors later reported that about half of these<br />

patients were in or near remission after three years of follow up. No<br />

adverse effects were reported in this study.<br />

CONCLUSION: Summarizing the data of these three double-blind studies,<br />

we may conclude that minocycline may be beneficial in patients with<br />

rheumatoid arthritis, especially when given early in the disease course or in<br />

patients with a mild disease.<br />

*Table 9 is an all-inclusive listing of evidence from clinical studies with tetracyclines. Some studies (Haider et al.) have been included due to other FDA<br />

approved indications of these agents.<br />

Additional Evidence<br />

Dose Simplification: Limited clinical studies have evaluated the impact of the dosing frequency with<br />

tetracyclines, especially with respect to impact on the outcome of disease. One study by Dunbar-Jacob et<br />

al. evaluated compliance to oral therapies for pelvic inflammatory disease. 38 In the randomized trial, study<br />

subjects took an average of 70% of prescribed doses, took the two daily prescribed doses of doxycycline<br />

for less than half of their outpatient days, took an unscheduled drug holiday for almost 25% of their<br />

outpatient days, and took only 16.9% of their doses within the optimal timing interval. These disturbing<br />

rates of adherence, even after hospitalization, suggest the need for education and antibiotic regimens<br />

involving shorter courses and longer dosing intervals, however, no conclusion on outcome of the diseasae<br />

was made. Another study by Lee et al. looked at the effectiveness of an enhanced compliance program for<br />

Helicobacter pylori therapy (treatment with bismuth subsalicylate, metronidazole, and tetracycline). 39 The<br />

enhanced group received medication counseling from a pharmacist, along with a medication calendar and a<br />

mini pillbox. There was no statistically significant difference between the groups in the number of patients<br />

taking more than 60% of the medications. However, there was a statistically significant difference in the<br />

number of patients taking more than 90% of the medications (67% of the control group vs. 89% of the<br />

enhanced compliance group; p


Impact on Physician Visits: A literature search of Medline and Ovid did not reveal clinical literature<br />

relevant to use of the tetracycline agents and their impact on physician visits.<br />

IX.<br />

Conclusions<br />

Since these medications are prescribed mostly because of sensitivity of organisms, all agents have a place<br />

in the treatment of infectious diseases. Several agents have specific uses for non-infectious disease states,<br />

such as demeclocycline for hyponatremia, and do not have appropriate substitutes as outlined in the<br />

comparison of efficacy studies listed above. Adverse reactions with the agents in this class are also<br />

comparable. Additionally, generic formulations are available for all oral tetracyclines in this class.<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the generics<br />

and offer no significant clinical advantage over other alternatives in general use.<br />

X. Recommendations<br />

No brand tetracycline is recommended for preferred status.<br />

218


References<br />

1. Bristol-Myers Squibb Company. Sumycin Syrup prescribing information. Princeton (NJ). May 2002.<br />

2. Bristol-Myers Squibb Company. Sumycin Tablets prescribing information. Princeton (NJ). July 2002.<br />

3. Roerig Pfizer. Terramycin Intramuscular Solution prescribing information. New York (NY). September<br />

2003.<br />

4. Roerig Pfizer. Vibramycin for Injection prescribing information. New York (NY). November 2001.<br />

5. Roerig Pfizer. Vibramycin and Vibra-Tabs prescribing information. New York (NY).September 2003.<br />

6. Lederle Pharmaceutical Division of American Cyanamid Company. Minocin Capsules prescribing<br />

information. Pearl River (NY). October 2003.<br />

7. Medicis. Dynacin prescribing information. Scottsdale (AZ). April 2003.<br />

8. Lederle Pharmaceutical Division of American Cyanamid Company. Declomycin Tablet prescribing<br />

information. Pearl River (NY). June 2003.<br />

9. Drug Facts and Comparisons 2004. Antiviral Agents [cited 2004 September 17] http://www.efactsweb.com<br />

10. Roberts MC. Tetracycline therapy: update. Clin Infect Dis. 2003 Feb 15;36(4):462-7. Epub 2003 Jan 28.<br />

11. Mullegger RR. Dermatological manifestations of Lyme borreliosis. Eur J Dermatol. 2004 Sep-<br />

Oct;12(5):296-309.<br />

12. Wormser GP, Ramanathan R, et al. Duration of antibiotic therapy for early Lyme disease. A randomized,<br />

double-blind, placebo-controlled trial. Ann Intern Med. 2003 May 6;138(9):697-704.<br />

13. Tilley BC, Alarcon GS, et al. Minocycline in rheumatoid arthritis. A 48-week, double-blind, placebocontrolled<br />

trial. MIRA Trial Group.<br />

14. Goh KP. Management of hyponatremia. Am Fam Physician. 2004 May 15;69(10):2387-94.<br />

15. Goulden V. Guidelines for the management of acne vulgaris in adolescents. Pediat Drugs. 2003;5(5):301-<br />

13.<br />

16. Garner SE, Eady EA, Popescu C, Newton J, Le WA. Minocycline for acne vulgaris: efficacy and safety.<br />

Cochrane Database Syst Rev. 2003;(1):CD002086.<br />

17. Haider A, Shaw J. Treatment of Acne Vulgaris. JAMA 2004 August 11;292(6):reprint.<br />

18. Saikali, Zeina, Sing, Gurmit. Doxycycline and other tetracyclines in the treatment of bone metastasis.<br />

Anti-Cancer Drugs. 2003 November;14(10):773-778.<br />

19. Shaprio L, Knowles S, Shear N. Comparative safety of tetracycline, minocycline and doxycycline.<br />

Archives of Dermatology. October 1997;133(10):1224-1230.<br />

20. Langevitz P, Livneh A, Bank I, Pras M. Benefits and Risks of minocycline in rheumatoid arthritis. Drug<br />

Safety 2000;22(5):405-414.<br />

21. Eichenfield A. Minocycline and autoimmunity. Current opinion in pediatrics. October 1999;11(5):477.<br />

22. O’Dell J, Paulsen G, Haire C, et al. Treatment of early seropositive rheumatoid arthritis with minocycline:<br />

Four year follow up of a double blind, placebo controlled trial. Arthritis and Rheumatism. 1999<br />

August;42(8):1691-1695.<br />

23. O’Dell J, Haire C, Palmer W, et al. Treatment of early rheumatoid arthritis with minocycline or placebo:<br />

Results of randomized, double-blind, placebo-controlled trial. Arthritis and Rheumatis. 1997<br />

May;40(5):842-848.<br />

24. Smilack JD. The tetracyclines. Mayo Clin Proc. 1999 July;74(7):727-9.<br />

25. Klein NC, Cunha BA. New uses of older antibiotics. Med Clin North Am. 2001 Jan;85(1):125-32.<br />

26. Stone M, Fortin PR, Pacheco-Tena C, Inman RD. Should tetracycline treatment be used more extensively<br />

for rheumatoid arthritis Mataanalysis demonstrates clinical benefit with reduction in disease activity. J<br />

Rheumatol. 2003 Oct;30(10):2112-22.<br />

27. Kinzie BJ. Management of the syndrom of inappropriate secretion of antidiuretic hormone. Clin Pharm.<br />

1987 Aug;6(8):625-33.<br />

28. Maesen FP, Davies BI, vanden Bergh JJ. Doxycycline and minocycline in the treatment of respiratory<br />

infections: a double-blind, comparative clinical, microbiological and pharmacokinetic study. J Antimicrob<br />

Chemother. 1989 Jan;23(1):123-9.<br />

29. Forrest JC, Cox M, Hon C, Morrison G, Bia N, Singer I. Superiority of democlocycline over lithium in the<br />

treatment of chronic syndrome of inappropriate secretion of antidiuretic hormone. N Engl J Med. 1978 Jan<br />

26;298(4):173-7.<br />

30. Kovacs GT, et al. A prospective single-blind trial of minocycline and doxycycline in the treatment of<br />

genital chalmydia trachomatis infection in women. Med J Aust. 1989 May 1;150(9):483-5.<br />

219


31. Romanowski B, et al. Minocycline compared with doxycycline in the treatment of nongonococcal urethritis<br />

and mucopurulent cervicitis. Ann of Int Med. 1993 July:119(1):16-22.<br />

32. Cunha B. Minocycline versus doxycycline in the treatment of Lyme neuroborreliosis. Clin Inf Diseases.<br />

200;30:237-238.<br />

33. Hanes PJ, Purvis JP. Local anti-infective therapy: pharmacological agents. A systematic review. Ann<br />

Periodontol. 2003 Dec;8(1):79-98.<br />

34. Klein NC, Cunha BA. Tetracyclines. Med Clin North Am. 1995 Jul;79(4(:789-801.<br />

35. Barza M, Schiefe RT. Antimicrobial spectrum, pharmacology and therapeutic use of antibiotics. Part 1:<br />

tetracyclines. Am J Hosp Pharm. 1977 Jan;24(1):49-57.<br />

36. Sum PE, Sum JW, Projan SJ. Recent developments in tetracycline antibiotics. Curr Pharm Des. 1998<br />

Apr;4(2):119-32.<br />

37. CDC. www.cdc.gov. Accessed 9/20/04.<br />

38. Dunbar-Jacob J, Sereika SM, foley SM, et al. Adherence to oral therapies in pelvic inflammatory disease.<br />

J Womens Health (Larchmt) 2004 Apr;13(3):285-91.<br />

39. Lee M, Kemp JA, Canning A, et al. A randomized controlled trial of an enhanced patient compliance<br />

program for Helicobacter pylori therapy. Arch Int Med 1999 Oct 25;159(19):2312-6.<br />

220


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of Nucleoside and Nucleotide Antiviral Agents<br />

AHFS 081832<br />

October 27, 2004<br />

I. Overview<br />

Herpesviruses<br />

There are eight identified human herpesviruses (HHVs) which are divided into three groups:<br />

• the alpha-herpesviruses (e.g., herpes simplex virus types 1 and 2 [HSV-1 and HSV-2]<br />

and varicella-zoster)<br />

• beta-herpesviruses (e.g., cytomegalovirus, HHV-6, and HHV-7)<br />

• gamma-herpesviruses (e.g., Epstein-Barr virus, Kaposi’s sarcoma-associated herpes<br />

virus, and HHV-8) 1<br />

The HHVs are all structurally similar with an internal core containing viral DNA, a capsid,<br />

and a lipid envelope. However, the HHVs are distinct with respect to their biologic and<br />

epidemiologic characteristics.<br />

Herpes Simplex Virus (HSV-1 and HSV-2)<br />

Herpes simplex virus infection can produce a multitude of illnesses including mucocutaneous infections,<br />

central nervous system infections, and infections of visceral organs. The two most common cutaneous<br />

manifestations of HSV are orolabial and genital lesions. The HSV-1 strain is most often associated with<br />

oropharyngeal disease and HSV-2 is most often associated with genital disease; however, both viruses can<br />

cause clinically indistinguishable infections in either of these anatomic areas. Genital herpes is one of the<br />

most common viral sexually transmitted diseases in the world, and it is estimated that at least 45 million<br />

people in the United States have genital HSV infection. 2 The most frequent mode of transmission is<br />

through direct contact of mucus membranes or abraded skin with infected secretions (e.g., oral, genital,<br />

mucosal) of someone who is shedding the virus. Initial infection may result in clinical manifestations (e.g.,<br />

lesions), but is often subclinical and asymptomatic, which may represent the most common source of<br />

transmission. 1,2 After viral inoculation, HSV replicates and spreads to peripheral sensory nerves. The virus<br />

remains latent in sensory or autonomic nerve root ganglia, with a lifelong potential for reactivation and<br />

recurrence. The factors involved in maintaining latency have not been fully elucidated; however, immune<br />

function and emotional and physical stressors appear to play an important role in reactivation of the virus.<br />

Additionally, immunocompromised patients often have more frequent and more severe episodes of<br />

reactivation. A diagnosis of HSV infection can often be made based on the presence of characteristic<br />

vesicular lesions; however, laboratory testing is necessary for confirmation. Herpes simplex virus can be<br />

isolated in a tissue culture or detected by the presence of HSV antigens or DNA in lesion samples.<br />

Varicella-Zoster Virus<br />

Another HHV, varicella-zoster virus (VZV) is responsible for two diseases: chickenpox and herpes zoster. 3<br />

Chickenpox, the primary infection, is highly contagious and is assumed to spread via the respiratory route.<br />

However, chickenpox is normally a benign disease characterized by a generalized exanthematous rash,<br />

low-grade fever, and malaise. Over 90% of chickenpox cases occur in children under the age of 13, and<br />

patients are contagious from approximately 48 hours before vesicles begin to form until all the vesicles<br />

have crusted. Mortality associated with chickenpox occurs in less than 2 out of every 100,000 cases;<br />

however, adults often have a more severe disease and risk of mortality is increased more than 15-fold. A<br />

live, attenuated virus vaccine for the prevention of chickenpox is available and recommended for normal<br />

children and in susceptible adults. After primary infection with chickenpox, VZV becomes latent within<br />

the dorsal root ganglia. Reactivation of VZV occurs sporadically and results in a herpes zoster infection<br />

(e.g., shingles), which is characterized by a unilateral vesicular eruption with a dermatomal distribution.<br />

Herpes zoster most commonly affects elderly patients; however, it can occur at any age and will occur in<br />

221


approximately 20% of the population at some point. Neuritis, ophthalmic complications, and postherpetic<br />

neuralgia are some of the most significant manifestations of herpes zoster. The mechanism responsible for<br />

reactivation is not known, but is likely dependent on both virus and host factors. Immunocompromised<br />

patients have a greater incidence of both chickenpox and shingles and often have greater disease severity as<br />

a result of infection. The diagnosis of both chickenpox and shingles is usually based on a history and<br />

physical examination; however, in unclear cases, VZV can be isolated from a tissue culture or serum<br />

antibody assays can be utilized.<br />

Cytomegalovirus<br />

Although cytomegalovirus (CMV) is highly prevalent, reaching 60% to 70% in urban cities of the United<br />

States, it rarely produces symptoms in patients with normal immune function. 4 Cytomegalovirus is the<br />

most common opportunistic pathogen in immunocompromised patients and produces the most severe<br />

symptoms in transplant patients. Similar to the other herpesviruses, CMV can become latent after primary<br />

infection. As with the other herpes viruses, the mechanism responsible for reactivation is unknown, but can<br />

occur as a result of immunosuppression, occurrence of other illness, or the use of chemotherapeutic<br />

medications. A diagnosis of CMV almost always requires laboratory confirmation and is based on growth<br />

of the virus from body fluids or the isolation of viral antigens or viral DNA. Complications of CMV<br />

infection include interstitial pneumonia, hepatitis, Guillain-Barre Syndrome, meningoencephalitis,<br />

myocarditis, colitis and esophageal ulcers, thrombocytopenia, hemolytic anemia, and skin eruptions. In<br />

patients with AIDS, CMV retinitis is the most common form of CMV disease, usually reported when the<br />

CD 4 cell count is less than 50 cells/mm 3 , and can lead to blindness.<br />

Chronic Hepatitis<br />

Chronic Hepatitis B<br />

An estimated 350 million people are chronically infected with Hepatitis B virus (HBV) worldwide, with<br />

1.25 million cases in the United States. 5,6 Hepatitis B infection can be self-limited or a chronic disease.<br />

Men who have sex with men, intravenous drug users, and people with multiple sex partners have an<br />

increased prevalence of HBV infection. Transmission of HBV occurs by percutaneous or mucous<br />

membrane exposure to infected body fluids, and sexual transmission among adults accounts for most cases<br />

in the United States. The development of chronic HBV infection following acute exposure is estimated at<br />

90% in newborns of mothers positive for hepatitis B e antigen (HBeAg), 25% to 30% in infants and<br />

children under the age of five, and less than 10% in adults. Individuals with compromised immune function<br />

are also at an increased risk of developing chronic HBV infection following acute exposure. Patients with<br />

chronic HBV are more likely to develop cirrhosis, hepatic decompensation, and hepatocellular carcinoma<br />

(HCC), although only 15% to 40% will develop serious manifestations during their lifetime. Additional<br />

risk factors for the development of HCC in patients with chronic HCV infection include male gender,<br />

family history of HCC, increased age, presence of HBeAg, history of reversions from anti-Hbe to HBeAg,<br />

cirrhosis, and coinfection with HCV.<br />

Chronic Hepatitis C<br />

Current estimates suggest that more than 2.7 million Americans have chronic HCV infection. 7,8 Studies<br />

indicate that 55% to 85% of persons who contract acute HCV will develop a chronic HCV infection.<br />

Younger age, female gender, and normal immune function are some factors that have been identified which<br />

may increase the likelihood of clearing an acute HCV infection. Chronic HCV infection is diagnosed by<br />

the detection of HCV RNA in the blood for at least six months. The most serious sequelae of HCV include<br />

liver fibrosis, cirrhosis, end-stage liver disease, and hepatocellular carcinoma (HCC), with an estimated 5%<br />

to 20% of patients developing cirrhosis over a period of 20 years. Hepatitis C virus represents the leading<br />

cause of death from liver disease in the United States. There are six genotypes of HCV, and in the United<br />

States genotype 1 accounts for 70% to 75% of HCV infections. Genotype 1 has also been associated with<br />

lower response to therapy. Transmission of HCV primarily occurs through contact with infected blood or<br />

blood products. Intravenous drug use is the leading route of transmission in the United States; however,<br />

blood transfusions prior to 1992, solid organ transplants from infected donors, occupational exposure to<br />

infected blood, birth to an HCV-positive mother, and multiple sexual partners are also potential sources of<br />

exposure. The goal of treatment is to prevent long-term complications of HCV infection, for which<br />

222


suppression of HCV RNA is used as a surrogate marker. Infection is considered eradicated when there is a<br />

sustained virologic response, defined as the absence of HCV RNA in serum at the end of treatment and six<br />

months post-therapy.<br />

Complications of chronic HBV or HCV may take decades to appear; therefore, virologic suppression or<br />

clearance and normalization of liver enzymes are often used to guide therapy and are used as surrogate<br />

markers for decreased risk of cirrhosis, HCC, or death.<br />

Respiratory Syncytial Virus<br />

Respiratory Syncytial Virus (RSV) is a common childhood disease, and almost all children will have been<br />

exposed to RSV by the age of two. 9 It is estimated that RSV infections account for 100,000<br />

hospitalizations in the United States each year. Most patients with RSV will experience only mild common<br />

cold symptoms; however, in vulnerable patients, RSV can progress to the lower respiratory tract causing<br />

bronchiolitis or pneumonia, with an increased risk of morbidity and mortality. Palivizumab (Synagis ® ) is a<br />

monoclonal antibody that is used to prevent the development of lower respiratory tract infection from RSV<br />

in high-risk infants (eg, premature, immunocompromised). Symptoms of RSV lower respiratory tract<br />

infection include tachypnea, wheezing, rales, hypoxemia, and apnea. Immunofluorescence or enzyme<br />

immunoassay rapid detection tests should be used to confirm a diagnosis of RSV infection. Patients with<br />

upper respiratory tract infections typically only require symptomatic therapy (eg, hydration, treatment of<br />

fever, management of nasal congestion). Treatment options for RSV lower respiratory infection include<br />

intravenous hydration, oxygen supplementation, bronchodilators, aerosolized ribavirin, and mucolytic<br />

therapy. Additionally, in the face of respiratory failure, corticosteroids, mechanical ventilation, and<br />

exogenous surfactant are considerations.<br />

The antiviral nucleoside and nucleotide analogs included in this review are summarized in Table 1. This<br />

review encompasses all systemic dosage forms and strengths.<br />

Table 1. Antiviral Nucleosides and Nucleotides in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name (s)<br />

Ganciclovir Oral Capsule, Powder for Injection Cytovene ® *, Cytovene ® -IV<br />

Valganciclovir Oral Tablet Valcyte <br />

Acyclovir Oral Tablet, Oral Capsule, Oral Suspension,<br />

Zovirax ® *<br />

Powder for Injection, Solution for Injection<br />

Famciclovir Oral Tablet Famvir ®<br />

Valacyclovir Oral Caplet Valtrex ®<br />

Cidofovir Solution for Injection Vistide ®<br />

Ribavirin Oral Capsule, Oral Tablet, Powder for Aerosol Rebetol ® *, Copegus ® , Virazole ® , Ribasphere ®<br />

Reconstitution<br />

Adefovir Oral Tablet Hepsera ®<br />

*Generic oral formulation available.<br />

223


II.<br />

Current Treatment Guidelines<br />

Herpes Simplex Virus<br />

The Centers for Disease Control and Prevention (CDC) have made recommendations for the management<br />

of genital herpes. 10 These recommendations are summarized in Table 2.<br />

Table 2. Centers for Disease Control and Prevention Treatment Guidelines for Genital<br />

Herpes Simplex Virus Infections* 10<br />

• Systemic antiviral medications partially control the signs and symptoms of herpes episodes<br />

when used to treat first clinical episodes and recurrent episodes, or when used as daily<br />

suppressive therapy.<br />

• Topical antiviral agents confer minimal clinical benefit, and the use of these medications is<br />

not recommended.<br />

• Initial episode: one of the following regimens.<br />

Acyclovir 400mg orally TID for 7-10 days<br />

Acyclovir 200mg orally five times daily for 7-10 days<br />

Famciclovir 250mg orally TID for 7-10 days<br />

Valacyclovir 1000mg BID for 7-10 days<br />

Treatment may be extended if healing is incomplete after 10 days<br />

• Recurrent episodes: one of the following regimens.<br />

Acyclovir 400mg orally TID for 5 days<br />

Acyclovir 200mg orally five times daily for 5 days<br />

Acyclovir 800mg orally BID for 5 days<br />

Famciclovir 125mg orally BID for 5 days<br />

Valacyclovir 500mg orally BID for 3-5 days<br />

Valacyclovir 1000mg orally QD for 5 days<br />

• Suppressive therapy for recurrent genital herpes: one of the following regimens.<br />

Acyclovir 400mg orally BID<br />

Famciclovir 250mg orally BID<br />

Valacyclovir 500mg orally QD<br />

Valacyclovir 1000mg orally QD<br />

• Severe disease: Intravenous acyclovir therapy should be provided for patients with severe<br />

disease or complications that require hospitalization, such as disseminated infection,<br />

pneumonitis, hepatitis, or complications of the central nervous system. The recommended<br />

regimen is intravenous acyclovir 5-10mg/kg body weight every 8 hours for 2-7 days or until<br />

clinical improvement is seen.<br />

• Treatment of Patients with HIV: one of the following regimens.<br />

Acyclovir 400mg TID for 5-10 days<br />

Acyclovir 200mg five times daily for 5-10 days<br />

Famciclovir 500mg BID for 5-10 days<br />

Valacyclovir 1000mg BID for 5-10 days<br />

• Suppressive Therapy in Patients with HIV: one of the following regimens.<br />

Acyclovir 400-800mg BID to TID<br />

Famciclovir 500mg BID<br />

Valacyclovir 500mg BID<br />

• Counseling of infected patients and their sex partners is critical to the management of genital<br />

herpes. Counseling should help patients cope with the infection and prevent sexual and<br />

perinatal transmission. The misconception that herpes simplex virus causes cancer should be<br />

dispelled, because the role of herpes virus in cervical cancer is at most that of a cofactor, not a<br />

primary etiologic agent.<br />

*TID indicates three times daily; BID, twice daily; QD, once daily.<br />

224


Varicella-Zoster Virus<br />

The American Academy of Pediatrics (AAP) does not recommend the use of oral acyclovir for treatment of<br />

varicella in otherwise healthy children. 11 Oral acyclovir should be considered for otherwise healthy<br />

patients at an increased risk for moderate to severe varicella infection, including people over 12 years of<br />

age, people with chronic cutaneous or pulmonary disorders, people receiving long-term salicylate therapy,<br />

and people receiving short, intermittent, or aerosolized courses of corticosteroids. Intravenous acyclovir is<br />

recommended for immunocompromised patients. The AAP does not have a recommendation regarding<br />

famciclovir and valacyclovir because pediatric formulations are not available and data on the use of these<br />

medications are lacking. No formal treatment guidelines for the management of herpes zoster infections<br />

were identified. 12 However, goals of therapy should include acceleration of healing, limitation of the<br />

intensity and duration of acute and chronic pain, and reduction of complications. For immunocompromised<br />

patients and those at an increased risk of more severe disease, treatment objectives should also include<br />

reducing the risk of VZV dissemination. Acyclovir, valacyclovir, and famciclovir have demonstrated<br />

benefit with respect to these treatment goals and are specifically indicated for the treatment of herpes<br />

zoster. 13-15<br />

Cytomegalovirus<br />

The United States Public Health Service in conjunction with the Infectious Disease Society of<br />

America (IDSA) has established guidelines for the prevention of opportunistic infections among<br />

HIV-infected patients. 16 For primary prevention of disease, these guidelines recommend consideration of<br />

prophylaxis with oral ganciclovir in HIV-infected adults and adolescents who are CMV-positive and have a<br />

CD 4 cell count


Table 3. American Association for the Study of Liver Diseases Recommendations for the Treatment<br />

of Chronic Hepatitis B Infection 5,6 *<br />

Presence of<br />

HBeAg<br />

Presence of<br />

HBV DNA† ALT Level Recommended Treatment<br />

Yes Yes ≤2 x ULN Observation only. Consider treatment when ALT becomes elevated.<br />

Yes Yes >2 x ULN • IFN-α for 16 weeks OR<br />

• Lamivudine for minimum of 1 year, continue 3-6 months after<br />

HBeAg seroconversion OR<br />

• Adefovir for minimum of 1 year<br />

• If nonresponder with IFN-α or contraindication, use lamivudine or<br />

adefovir<br />

• Adefovir in patients with lamivudine resistance.<br />

No Yes >2 x ULN IFN-α, lamivudine, or adefovir can be used; however, IFN-α or adefovir<br />

is preferred due to need for long-term therapy.<br />

• IFN-α for 16 weeks OR<br />

• Adefovir for minimum of 1 year OR<br />

• Lamivudine for minimum of 1 year, continue 3-6 months after<br />

HBeAg seroconversion<br />

• If nonresponder with IFN-α or contraindication, use lamivudine or<br />

adefovir<br />

• Adefovir in patients with lamivudine resistance.<br />

No No ≤ x ULN Observation only.<br />

Yes or No Yes Cirrhosis • Compensated cirrhosis: lamivudine or adefovir<br />

• Decompensated cirrhosis: lamivudine or adefovir; refer for liver<br />

transplant; IFN-α is contraindicated.<br />

Yes or No No Cirrhosis • Compensated: observation only.<br />

• Decompensated: refer for liver transplant.<br />

*HBeAg indicates hepatitis B e antigen; HBV, hepatitis B virus; ALT, alanine aminotransferase; ULN, upper limit of normal; IFN-α,<br />

interferon-alfa.<br />

†Presence indicates HBV DNA >10 5 copies/mL, which was arbitrarily chosen.<br />

Interferon, ribavirin, or a combination of interferon and ribavirin are indicated for the treatment of chronic<br />

HCV infection. 7 Recent guidelines issued by the American Association for the Study of Liver Diseases for<br />

the treatment of chronic HCV infection recommend initiation of therapy in patients 18 years of age and<br />

older, who have abnormal ALT levels, liver biopsy demonstrating significant fibrosis, compensated liver<br />

disease, and acceptable hematological and biochemical indices (e.g., hemoglobin >13g/dL for men and<br />

>12g/dL for women, creatinine 75kg.<br />

Genotype-2 or Genotype-3 HCV Infection<br />

• Peginterferon plus ribavirin for 24 weeks<br />

• Ribavirin dose = 800mg<br />

Genotypes 4, 5, and 6 HCV Infection<br />

• Insufficient data to provide recommendations<br />

Respiratory Syncytial Virus<br />

Recommendations by the AAP for treatment of RSV state that primary treatment is supportive and should<br />

include hydration, monitoring of respiratory status, supplemental oxygen, and mechanical ventilation if<br />

necessary. 21 Although aerosolized ribavirin has demonstrated good in vitro activity against RSV, an<br />

inconsistent demonstration of clinical benefit has been observed in placebo-controlled trials. Therefore,<br />

the AAP does not generally recommend the use of aerosolized ribavirin for treatment of RSV infection. In<br />

addition to conflicting results from clinical trials, the cost of aerosolized ribavirin as well as concerns about<br />

226


potential toxic effects to exposed health care professionals, have contributed to this recommendation. The<br />

AAP states that decisions regarding administration of aerosolized ribavirin should be made based on<br />

individual clinical circumstances.<br />

227


III. Comparative Indications of the Antiviral Nucleosides and Nucleotides<br />

Table 4 lists the FDA-approved indications for the antiviral agents included in this review.<br />

Table 4. FDA-Approved Indications for the Nucleoside and Nucleotide Agents* 13-15,17,18,20,22-27<br />

Drug<br />

CMV<br />

Retinitis<br />

Prevention<br />

of CMV<br />

Disease<br />

Mucocutaneous<br />

HSV Infections<br />

Initial<br />

Genital<br />

Herpes<br />

Episodes<br />

Recurrent<br />

Genital<br />

Herpes<br />

Episodes<br />

Chronic<br />

Suppression<br />

of Recurrent<br />

Genital<br />

Herpes<br />

HSV<br />

Encephalitis<br />

Herpes<br />

Zoster<br />

(Shingles)<br />

Chickenpox<br />

Chronic<br />

Hepatitis<br />

C<br />

Chronic<br />

Hepatit<br />

s B<br />

Lower<br />

Respiratory<br />

Infection<br />

caused by<br />

RSV<br />

Oral Ganciclovir X†‡ X†<br />

IV Ganciclovir X§ X║<br />

Valganciclovir X X║<br />

Cidofovir X<br />

Oral Acyclovir X X X X X<br />

IV Acyclovir X§ X** X X§ X§<br />

Famciclovir X†† X** X** X<br />

Valacyclovir X X** X** X**†† X<br />

Oral Ribavirin X‡‡<br />

Aerosolized<br />

Ribavirin X<br />

Adefovir X<br />

*CMV indicates cytomegalovirus; HSV, herpes simplex virus; RSV, respiratory syncytial virus; IV, intravenous. Boxes marked with "X" designate an indication approved by the Food and Drug Administration.<br />

†In immunocompromised patients, including patients with advanced HIV infection or transplant patients.<br />

‡For maintenance treatment in patients with stable retinitis following induction therapy.<br />

§In immunocompromised patients.<br />

║In transplant recipients.<br />

In patients with AIDS.<br />

**In patients with normal immune function.<br />

††In patients with HIV.<br />

‡‡Interferon must be given with ribavirin. Rebetol ® is indicated in combination with interferon alfa-2b, recombinant or peginterferon alfa-2b, recombinant. Copegus ® is indicated in combination with peginterferon<br />

alfa-2a only.<br />

228


IV.<br />

Pharmacokinetic Parameters of the Antiviral Nucleosides and Nucleotides<br />

Table 5 lists the pharmacokinetic parameters and mechanisms of action of these antiviral agents.<br />

Table 5. Pharmacokinetic Parameters of Antiviral Nucleoside and Nucleotide Agents 13-15,17,18,20,22-27<br />

Drug<br />

Mechanism of<br />

Action Bioavailability<br />

Protein<br />

Binding Metabolism<br />

Active<br />

Metabolites Elimination<br />

Ganciclovir Synthetic Poorly 1%-2% CMV-encoded<br />

guanine absorbed<br />

protein kinase<br />

nucleoside orally (5%<br />

and other<br />

analog. fasting, 6%-<br />

cellular kinases<br />

Converted by a 9% with<br />

CMV encoded food).<br />

protein kinase to Intravenous<br />

ganciclovir formulation<br />

monophosphate, available.<br />

that is further<br />

phosphorylated<br />

to ganciclovir<br />

triphosphate by<br />

cellular kinases.<br />

The ganciclovir<br />

triphosphate<br />

inhibits viral<br />

DNA synthesis<br />

and replication of<br />

herpes viruses<br />

either by<br />

competitive<br />

inhibition of<br />

DNA<br />

polymerases or<br />

direct<br />

incorporation<br />

into viral DNA.<br />

Valganciclovir<br />

Cidofovir<br />

Prodrug of<br />

ganciclovir.<br />

Rapidly<br />

converted to<br />

ganciclovir by<br />

intestinal and<br />

hepatic esterases.<br />

Nucleotide<br />

analog that<br />

suppresses CMV<br />

replication by<br />

inhibition of viral<br />

DNA syntheses.<br />

Cidofovir is<br />

phosphorylated<br />

into active<br />

cidofovir<br />

diphosphate that<br />

inhibits DNA<br />

polymerase and<br />

incorporates into<br />

viral DNA.<br />

60% with food 1%-2% Intestinal and<br />

hepatic<br />

esterases (once<br />

converted to<br />

ganciclovir,<br />

also undergoes<br />

metabolism by<br />

CMV encoded<br />

protein kinase<br />

and other<br />

cellular kinases)<br />

Intravenous<br />

formulation<br />

only.<br />

Half-<br />

Life<br />

Yes Renal Oral:<br />

4.8<br />

hours<br />

IV: 3.5<br />

hours<br />

Yes Renal 4.1<br />

hours<br />


Acyclovir<br />

Famciclovir<br />

Valacyclovir<br />

Ribavirin<br />

Synthetic purine<br />

nucleoside<br />

analog.<br />

Converted by<br />

thymidine kinase<br />

encoded by HSV<br />

and VZV into<br />

acyclovir<br />

monophosphate<br />

that is further<br />

phosphorylated<br />

by other enzymes<br />

into acyclovir<br />

triphosphate.<br />

Acyclovir<br />

triphosphate<br />

inhibits viral<br />

DNA replication<br />

by competitive<br />

inhibition of viral<br />

DNA<br />

polymerase,<br />

incorporation<br />

into viral DNA,<br />

and inactivation<br />

of viral DNA<br />

polymerase.<br />

Rapid<br />

biotransformation<br />

to penciclovir,<br />

which is<br />

phosphorylated<br />

by thymidine<br />

kinase to<br />

penciclovir<br />

monophosphate,<br />

that is then<br />

converted to<br />

penciclovir<br />

triphosphate.<br />

Penciclovir<br />

triphosphate<br />

inhibits viral<br />

DNA synthesis<br />

and replication<br />

via competitive<br />

inhibition of<br />

HSV-2<br />

polymerase.<br />

Prodrug of<br />

acyclovir.<br />

Rapidly<br />

converted to<br />

acyclovir by<br />

first-pass<br />

intestinal and/or<br />

hepatic<br />

metabolism.<br />

Unknown. May<br />

act as an analog<br />

of guanosine or<br />

10%-20%,<br />

which<br />

decreases with<br />

increasing<br />

doses.<br />

Intravenous<br />

formulation<br />

also available.<br />

9%-33% Thymidine<br />

kinase and other<br />

enzymes<br />

77%


Adefovir<br />

xanthosine.<br />

Nucleotide<br />

analog that<br />

inhibits HBV<br />

DNA polymerase<br />

via competitive<br />

inhibition of<br />

binding site and<br />

incorporation<br />

into viral DNA.<br />

Phosphorylated<br />

into active<br />

adefovir<br />

diphosphate.<br />

is absorbed<br />

systemically.<br />

deribosylation/<br />

amide<br />

hydrolysis<br />

59% ≤4% Phosphorylation<br />

by cellular<br />

kinases.<br />

urine; 12%<br />

of oral dose<br />

is excreted<br />

in feces.<br />

Aerosol:<br />

9.5<br />

hours<br />

Yes Renal 7.5<br />

hours<br />

231


V. Drug Interactions of the Antiviral Nucleosides and Nucleotides<br />

Drug interactions identified for the nucleoside and nucleotide antiviral medications in this review are<br />

summarized below.<br />

Table 6. Drug Interactions of the Antiviral Nucleoside and Nucleotide Agents 28<br />

Drug Significance Interaction Mechanism<br />

Ganciclovir Level 3 Ganciclovir and didanosine Mechanism is unknown. Didanosine concentrations<br />

are increased during coadministration with<br />

ganciclovir, possibly resulting in didanosine<br />

toxicity. Because both ganciclovir and didanosine<br />

are partially eliminated via renal secretion, this<br />

interaction could be the result of competitive<br />

inhibition of renal secretion.<br />

Ganciclovir Level 1 Ganciclovir and zidovudine Mechanism is unknown. Combination therapy<br />

with ganciclovir and zidovudine in the<br />

treatment of cytomegalovirus disease<br />

increases hematological toxicity. The toxicity<br />

appears to be related to combined<br />

pharmacodynamic effects rather than to a<br />

pharmacokinetic interaction. This<br />

combination should be avoided until<br />

additional information is available.<br />

Acyclovir Level 4 Acyclovir and cimetidine Cimetidine reduces the rate but not extent of<br />

conversion of valcyclovir to acyclovir. Acyclovir<br />

renal clearance is reduced by concomitant<br />

administration of cimetidine. Cimetidine moderately<br />

increases acyclovir concentrations; however, the<br />

clinical significance of this interaction is probably<br />

limited.<br />

Acyclovir Level 4 Acyclovir and probenecid Acyclovir renal clearance is reduced by<br />

concomitant probenacid administration. Probenacid<br />

increases acyclovir concentrations moderately;<br />

however, the clinical significance of this interaction<br />

is probably limited.<br />

Acyclovir Level 4 Acyclovir and zidovudine Mechanism is unknown. A single case of severe<br />

lethargy and fatigue has been reported following the<br />

coadministration of acyclovir and zidovudine.<br />

Acyclovir does not appear to affect zidovudine<br />

concentrations.<br />

Famciclovir Level 4 Famciclovir and digoxin Mechanism is unknown. Famciclovir administration<br />

produces a small increase in digoxin peak plasma<br />

concentrations; however, this change is not likely to<br />

enhance patient response to digoxin.<br />

Ribavirin Level 3 Ribavirin and warfarin Mechanism is unknown. One patient on warfarin<br />

developed reduced anticoagulant effect when<br />

ribavirin was added. Although a causal relationship<br />

was evident in this case, it is not known how often<br />

other patients would be similarly affected.<br />

Anticoagulant effect should be monitored if<br />

ribavirin is initiated, discontinued, or changed with<br />

respect to dosage.<br />

*Level 1 indicates an interaction where risks always outweigh benefit of therapy and combination should be avoided; Level 2, use combination only under<br />

special circumstances; Level 3, take action as necessary to reduce risk; Level 4, risk of adverse outcomes appears small and no action is needed.<br />

232


Other interactions (per manufacturers labeling):<br />

Ganciclovir 17<br />

• Ganciclovir and probenecid = Probenecid 500mg every 6 hours increased ganciclovir (1000mg<br />

every 8 hours) area under the concentration-time curve (AUC) 53% ± 91% (range, -14% to<br />

299%). Renal clearance of ganciclovir decreased 22 ± 20% (range, -54% to –4%) indicating an<br />

interaction involving competition for renal tubular secretion.<br />

• Ganciclovir and imipenem-cilastatin = Generalized seizures have been reported in patients who<br />

received concomitant ganciclovir and imipenem-cilastatin. These drugs should not be used<br />

concomitantly unless the potential benefits outweigh the risks.<br />

• It is possible that drugs that inhibit replication of rapidly dividing cells may have additive toxicity<br />

when administered with ganciclovir. Therefore, drugs such as dapsone, pentamidine, flucytosine,<br />

vincristine, vinblastine, adriamycin, amphotericin B, trimethoprim/sulfamethoxazole, or other<br />

nucleoside analogues, should be considered for concomitant use with ganciclovir only if the<br />

potential benefits outweigh the potential risks.<br />

Valganciclovir 20<br />

• No in vivo evaluations of drug-drug interactions were conducted with valganciclovir. However,<br />

because it is a prodrug of ganciclovir and is rapidly and extensively converted, interactions<br />

associated with ganciclovir would be expected with valganciclovir.<br />

Valacyclovir 21<br />

• Valacyclovir and cimetidine = A single dose of cimetidine 800mg with valacyclovir 1000mg<br />

increased the AUC and maximum plasma concentration (C max ) of acyclovir by 32% and 8%,<br />

respectively. This effect is not thought to be clinically significant in patients with normal renal<br />

function.<br />

• Valacyclovir and probenacid = A single dose of probenecid 1000mg with valacyclovir 1000mg<br />

increased the AUC and maximum plasma concentration (C max ) of acyclovir by 49% and 22%,<br />

respectively. This effect is not thought to be clinically significant in patients with normal renal<br />

function.<br />

Famciclovir 15<br />

• Concomitant use of famciclovir and probenecid or other drugs significantly eliminated by active<br />

renal tubular secretion may result in increased plasma concentrations of penciclovir.<br />

Cidofovir 18<br />

• Cidofovir and probenecid = Concomitant administration of cidofovir and probenacid reduced the<br />

amount of cidofovir excreted unchanged in the urine to 70% to 85% of the dose. The renal<br />

clearance of cidofovir was reduced to a level consistent with creatinine clearance, indicating that<br />

probenecid blocks active renal tubular secretion of cidofovir. Probenecid must be administered<br />

with each cidofovir dose to minimize the risk of potential nephrotoxicity.<br />

• Cidofovir and nephrotoxic agents = Concomitant administration of cidofovir and agents with<br />

nephrotoxic potential is contraindicated.<br />

Adefovir 27<br />

• Adefovir and ibuprofen = Ibuprofen 800mg TID increased adefovir C max by 33% and AUC by<br />

23%. This interaction appears to be due to higher oral bioavailability, not a reduction in renal<br />

clearance of adefovir.<br />

Ribavirin 24,25<br />

• Ribavirin and didansoine = Exposure to didanosine is increased from concomitant administration<br />

with ribavirin, which could cause or worsen clinical toxicities. Coadministration of ribavirin and<br />

didanosine is not recommended.<br />

233


• Ribavirin and stavudine = Ribavirin has been shown in vitro to inhibit phosphorylation of<br />

stavudine, which could lead to decreased antiretroviral activity. Concomitant use of these<br />

medications should be used with caution.<br />

• Ribavirin and zidovudine = Ribavirin has been shown in vitro to inhibit phosphorylation of<br />

zidovudine, which could lead to decreased antiretroviral activity. Concomitant use of these<br />

medications should be used with caution.<br />

VI.<br />

Adverse Drug Events of the Nucleoside and Nucleotide Antiviral Agents<br />

Acyclovir, Valacyclovir, Famciclovir<br />

Adverse events for oral acyclovir, intravenous acyclovir, famciclovir, and valacylovir have been relatively<br />

similar in comparative clinical trials. Because valacyclovir is rapidly converted to acyclovir in vivo, a<br />

similar adverse event profile would be expected for these medications. Renal failure, in some cases<br />

resulting in death, has been observed with acyclovir and valacyclovir therapy. 13,14 Concomitant use of<br />

other nephrotoxic drugs, preexisting renal disease, and dehydration make renal impairment with acyclovir<br />

more likely. Intravenous acyclovir must be given over at least one hour to reduce the risk of renal tubular<br />

damage. 13 Thrombotic thrombocytopenic pupura/hemolytic uremic syndrome, which has resulted in death,<br />

has also occurred in immunocompromised patients receiving acyclovir and valacyclovir therapy. 13,14<br />

Additionally, approximately 1% of patients receiving intravenous acyclovir have experienced<br />

encephalopathic changes characterized by lethargy, obtundation, tremors, confusion, hallucinations,<br />

agitation, seizures, or coma. 13 The manufacturer recommends that acyclovir be used with caution in<br />

patients with underlying neurologic abnormalities or serious renal, hepatic, or electrolyte abnormalities, or<br />

significant hypoxia. Acute renal failure has also been reported in patients receiving famciclovir. 15<br />

Ganciclovir, Valganciclovir, Cidofovir<br />

In clinical trials comparing oral ganciclovir to intravenous ganciclovir, subjects treated with intravenous<br />

ganciclovir experienced lower minimum absolute neutrophil counts and hemoglobin levels, consistent with<br />

more neutropenia and anemia, compared to subjects treated with oral ganciclovir. 21 Retinal detachment has<br />

been reported in subjects with CMV retinitis, both before and after initiation of ganciclovir. 17 The<br />

relationship between retinal detachment and treatment with ganciclovir is unknown. The safety profiles of<br />

ganciclovir and valganciclovir have been similar in comparative clinical trials, which would be expected<br />

because valganciclovir is a prodrug of ganciclovir. Granulocytopenia (neutropenia), anemia, and<br />

thrombocytopenia have been observed in patients receiving ganciclovir or valganciclovir therapy 17,20 ;<br />

therefore, the manufacturer does not recommend the use of ganciclovir or valganciclovir in patients with an<br />

absolute neutrophil count


Adefovir<br />

Severe acute exacerbation of hepatitis has been reported in patients who have discontinued adefovir therapy<br />

for chronic HBV. 27 Nephrotoxicity, lactic acidosis, and severe hepatomegaly with steatosis, including fatal<br />

cases, have also been reported in patients receiving adefovir.<br />

Aerosolized Ribavirin<br />

Sudden deterioration of respiratory function has been associated with initiation of aerosolized ribavirin in<br />

infants. 26 Respiratory function needs to be carefully monitored during ribavirin therapy. Administration of<br />

aerosolized ribavirin to mechanically ventilated patients should only be undertaken by health care<br />

professionals experienced with this mode of administration and the specific ventilator being used.<br />

Attention must be paid to procedures that have been shown to minimize the accumulation of drug<br />

precipitate, which can result in mechanical ventilator dysfunction and associated increased pulmonary<br />

pressure. Additionally, ribavirin is teratogenic and studies have shown that the drug can disperse into<br />

surrounding bedside area, placing caretakers at risk. Other adverse events associated with aerosolized<br />

ribavirin include deaths during or shortly after treatment, some possibly related to ribavirin and some<br />

attributed to ribavirin precipitation in mechanical ventilation systems. Pulmonary and cardiovascular<br />

complications, anemia, rash, conjunctivitis, seizures, and asthenia have also been observed with aerosolized<br />

ribavirin therapy.<br />

Adverse events observed in clinical trials with these medications are listed in Table 7. However, this list<br />

may not include every adverse event reported in all clinical trials with these drugs. Additionally, as many<br />

of these agents are used in various patient populations (e.g., transplant recipients, HIV-infected individuals)<br />

and for different disease processes, adverse event percentages are often different between the respective<br />

studies depending on the patient population. Where noted, some percentages in the following Table may<br />

reflect an average of adverse event incidences from various trials. Please refer to the manufacturer labeling<br />

for additional safety information regarding specific disease states and treatment groups.<br />

235


Table 7. Common Adverse Events (%) Reported for the Antiviral Nucleosides and Nucleotides 13-15,17,18,20,22,23,27<br />

Adverse Event Ganciclovir* Valganciclovir† Cidofovir Acyclovir*‡ Famciclovir‡ Valacyclovir‡ Adefovir<br />

Body as a Whole<br />

Malaise<br />

Fever<br />

Chills<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Gastrointestinal<br />

Abdominal Pain<br />

Nausea<br />

Vomiting<br />

Diarrhea<br />

Anorexia<br />

Flatulence<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Depression<br />

Headache<br />

Neuropathy<br />

Paresthesia<br />

Insomnia<br />

Tremor<br />

Migraine<br />

Seizures<br />

Hepatic<br />

Increased LFTs<br />

Hepatitis<br />

Elevated total bilirubin<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Sweating<br />

Injection site reaction<br />

Urticaria<br />

Hematologic<br />

Neutropenia<br />

Leukopenia<br />

Anemia<br />

Thrombocytopenia<br />

Agranulocytosis<br />

Thrombocytosis<br />

Leukocytosis<br />

Neutrophilia<br />

Hemolytic anemia<br />

Genitourinary<br />

Increased SCr/BUN<br />

Acute renal failure<br />

Hematuria<br />

Dysmenorrhea<br />

Proteinuria<br />

Other<br />

Infection<br />

Sepsis<br />

Pneumonia<br />

Vitreous disorder<br />

Catheter-related infection<br />

Retinal detachment<br />

Graft rejection<br />

Decreased serum HCO 3<br />

Decreased intraocular pressure<br />

Uveitis<br />

Dyspnea<br />

<br />

38/48<br />

7/10<br />

<br />

<br />

<br />

17/19<br />

26/25<br />

13/13<br />

41/44<br />

15/14<br />

6/3<br />

<br />

<br />

<br />

8/9<br />

6/10<br />

<br />

<br />

<br />

31/13 58<br />

22<br />

<br />

<br />

-/18<br />

15/-<br />

30/23<br />

21/16<br />

41/30<br />

<br />

<br />

22/22<br />

9/-<br />

8/-<br />

16/20<br />

-/28<br />

<br />

<br />

<br />

<br />

69<br />

7<br />

26<br />

23<br />

<br />

<br />

<br />

30<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

15/10<br />

6/5<br />

11/12<br />

<br />

<br />

17/38<br />

29/41<br />

19/25<br />

6/6<br />

<br />

<br />

<br />

9/13<br />

4/15<br />

6/8<br />

6/4<br />

4/9<br />

8/11<br />

<br />

-/14<br />

27/-<br />

26/-<br />

6/-<br />

<br />

15/-<br />

-/24<br />

27<br />

30<br />

<br />

<br />

43<br />

<br />

24<br />

<br />

24<br />

<br />

<br />

88<br />

28<br />

9<br />

16<br />

24<br />

11<br />

23<br />

11.5/- 5.2<br />

3.3/7║<br />

3.3/7║<br />

2.8/-<br />


VII.<br />

Dosing and Administration of the Antiviral Nucleosides and Nucleotides<br />

Table 6. Dosing for the Antiviral Nucleoside and Nucleotide Agents 13-15,17,18,20,22-27<br />

Drug Availability Dose/Frequency/Duration<br />

Ganciclovir 250mg oral capsule;<br />

500mg oral capsule<br />

Dosing depends on indication. Capsules should be taken with food.<br />

500mg/10mL vial<br />

ganciclovir sodium for<br />

injection<br />

Treatment of CMV Retinitis<br />

Induction:<br />

• IV: 5mg/kg (at a constant rate over 1 hour) every 12 hours for 14-21 days.<br />

• Capsules are not indicated for induction.<br />

Maintenance:<br />

• IV: 5mg/kg (at a constant rate over 1 hour) QD, 7 days per week, or 6mg/kg<br />

QD, 5 days per week OR<br />

• Oral: 1000mg TID with food, or 500mg 6 times daily every 3 hours with<br />

food, during waking hours.<br />

Prevention of CMV Disease in Patients with Advanced HIV<br />

1000mg orally TID with food.<br />

Prevention of CMV Disease in Transplant Recipients<br />

• IV: initial dose of 5mg/kg (at a constant rate over 1 hour) every 12 hours for<br />

7-14 days, followed by 5mg/kg QD, 7 days per week or 6mg/kg QD, 5 days<br />

per week.<br />

• Oral: 1000mg TID with food.<br />

• Duration of treatment in transplant recipients depends on the duration and<br />

degree of immunosuppression.<br />

Other Notes:<br />

Ganciclovir should not be administered to a patient with an absolute neutrophil<br />

count


Acyclovir<br />

Famciclovir<br />

200mg capsule;<br />

400mg capsule;<br />

800mg capsule<br />

200mg/5mL suspension<br />

500mg/10mL and<br />

1000mg/10mL vials of<br />

acyclovir sodium for<br />

injection<br />

125mg tablets;<br />

250mg tablets;<br />

500mg tablets<br />

3 hours prior to cidofovir, and 1000mg at 2 and again at 8 hours after<br />

completion of cidofovir infusion).<br />

• Cidofovir is contraindicated in patients with a creatinine clearance<br />

≤55mL/min.<br />

Dosing depends on indication.<br />

Herpes Zoster<br />

• Oral: 800mg every 4 hours, 5 times daily for 7-10 days.<br />

Genital Herpes<br />

Initial episode:<br />

• Oral: 200mg every 4 hours, 5 times daily for 10 days OR<br />

• IV: 5mg/kg (at a constant rate over 1 hour), every 8 hours for 5 days.<br />

Chronic suppression for recurrent disease:<br />

• Oral: 400mg BID for up to 12 months, followed by re-evaluation. Other<br />

regimens have included doses ranging from 200 mg TID to 200mg 5 times<br />

daily.<br />

Intermittant therapy:<br />

• Oral: 200mg every 4 hours, 5 times daily for 5 days. Therapy should be<br />

initiated at the earliest sign of symptom of recurrence.<br />

Treatment of Chickenpox<br />

Therapy should be initiated at the earliest sign or symptom of chickenpox as<br />

there is no information about the efficacy of therapy initiated more than 24 hours<br />

after onset of signs and symptoms.<br />

Children:<br />

• Oral: 20mg/kg/dose QID for 5 days. Children over 40kg should receive the<br />

adult dose.<br />

Adults:<br />

• Oral: 800mg QID for 5 days.<br />

Immunocompromised children:<br />

• IV: 20mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 7 days.<br />

Immunocompromised adults:<br />

• IV: 10mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 7 days.<br />

Mucosal and Cutaneous HSV Infections in Immunocompromised Patients<br />

Adults:<br />

• IV: 5mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 7 days.<br />

Pediatrics:<br />

• IV: 10mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 7 days.<br />

Herpes Simplex Encephalitis<br />

Adults:<br />

• IV: 10mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 10 days.<br />

Pediatrics:<br />

• IV: 20mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 10 days.<br />

Neonatal HSV Infections (birth to 3 months):<br />

• IV: 10mg/kg/dose (at a constant rate over 1 hour) every 8 hours for 10 days.<br />

Doses of 15mg/kg or 20mg/kg at the same frequency have also been used;<br />

however, the safety and efficacy of these doses are not known.<br />

Dosage depends on indication.<br />

Herpes Zoster<br />

• 500mg every 8 hours for 7 days.<br />

238


Valacyclovir<br />

Ribavirin<br />

500mg caplets;<br />

1000mg caplets<br />

200mg tablet;<br />

200mg capsule<br />

• Therapy should be initiated as soon as herpes zoster is diagnosed, as no data<br />

are available on efficacy of treatment started greater than 72 hours after<br />

onset of rash.<br />

Genital Herpes<br />

Recurrent genital herpes:<br />

• 125mg BID for 5 days.<br />

• Therapy should be initiated at the first sign or symptom if medical<br />

management is indicated, as efficacy has not been established when<br />

treatment is initiated more than 6 hours after onset of symptoms or lesions.<br />

Suppression of recurrent genital herpes:<br />

• 250mg BID for 5 days for up to 1 year.<br />

• The safety and efficacy of famciclovir therapy beyond 1 year of treatment<br />

have not been established.<br />

Recurrent Orolabial or Genital Herpes in HIV-Infected Patients<br />

• 500mg BID for 7 days.<br />

Dosage depends on indication.<br />

Herpes Zoster<br />

• 1000mg TID for 7 days.<br />

• Therapy should be initiated at earliest sign or symptom of herpes zoster, and<br />

is most effective when started within 48 hours of the onset of zoster rash.<br />

There are no data available on efficacy of treatment initiated greater than<br />

72 hours after onset of rash.<br />

Genital Herpes<br />

Initial episodes:<br />

• 1000mg BID for 10 days.<br />

• Therapy is most effective when initiated within 48 hours of the onset of<br />

signs and symptoms. There are no data on the efficacy of treatment when<br />

initiated more than 72 hours after the onset of signs and symptoms.<br />

Recurrent episodes:<br />

• 500mg BID for 3 days.<br />

• Therapy should be initiated at the first sign or symptom of an episode if<br />

medical management is indicated. There are no data on the efficacy of<br />

treatment when initiated more than 24 hours after the onset of signs or<br />

symptoms.<br />

Suppression of recurrent genital herpes:<br />

• 1000mg QD.<br />

• 500mg QD is an alternative for patients with 9 or fewer recurrences per year.<br />

Suppression of recurrent genital herpes in HIV-infected patients:<br />

• Patients with CD 4 cell count ≥100 cells/mm 3 may receive 500mg BID.<br />

Reduction of transmission:<br />

• 500mg QD for the source partner is recommended for reduction of<br />

transmission of genital herpes in patients with a history of 9 or fewer<br />

recurrences per year.<br />

Herpes Labialis (cold sores)<br />

• 2000mg BID for 1 day.<br />

• Therapy should be initiated at earliest symptom of a cold sore, as there are<br />

no data on the effectiveness of treatment initiated after the development of<br />

clinical signs of a cold sore (e.g., papules, vesicles, or ulcer).<br />

*Monotherapy is not effective for the treatment of chronic HCV infection.<br />

Ribavirin must be used in combination with interferon therapy. The safety<br />

239


40mg/mL oral solution<br />

6grams lyophilized<br />

ribavirin powder for<br />

reconstitution and<br />

inhalation<br />

and efficacy of ribavirin tablets (Copegus ® ) have only been established when<br />

used in combination with pegylated interferon alfa-2a, recombinant. The safety<br />

and efficacy of ribavirin capsules and oral solution (Rebetol ® ) have been<br />

established when used in combination with interferon alfa-2b or pegylated<br />

interferon alfa-2b.<br />

Treatment of Chronic Hepatitis C with Ribavirin (Copegus ® ) plus pegylated<br />

interferon alfa-2a, recombinant<br />

Genotypes 1 and 4:<br />

• 75kg should receive ribavirin 600mg (3 capsules) QAM and 600mg<br />

(3 capsules) QPM daily with interferon alfa-2b.<br />

• The recommended duration of treatment for previously untreated patients is<br />

24-48 weeks. In patients who relapse following non-pegylated interferon<br />

monotherapy, the recommended duration of treatment is 24 weeks.<br />

Pediatrics<br />

• 15mg/kg/day divided into an AM and PM dose.<br />

• The recommended duration of therapy is 48 weeks for pediatric patients with<br />

genotype 1, and 24 weeks for pediatric patients with genotype 2/3.<br />

Treatment of Chronic Hepatitis C with Ribavirin (Rebetol ® ) plus pegylated<br />

interferon alfa-2b, recombinant<br />

Adults:<br />

• Ribavirin 400mg (2 capsules) QAM and 400mg (2 capsules) QPM with<br />

peginterferon alfa-2b.<br />

Other Notes Regarding Oral Ribavirin<br />

• A dosage reduction to 600mg daily (200mg in the morning, two 200mg<br />

tablets in the evening) is recommended for certain decreases in hemoglobin<br />

(e.g.,


Adefovir 10mg tablet Chronic Hepatitis B Infection<br />

• 10mg QD.<br />

• The optimal duration of therapy is unknown.<br />

*Dosages listed in this table are based on normal renal function. Dosages should be adjusted according to manufacturer recommendations for patients<br />

with impaired renal function.<br />

Special Dosing Considerations<br />

Table 7. Special Dosing Considerations for the Nucleoside and Nucleotide Antiviral Agents 13-15,17,18,20,22-27<br />

Drug Renal Hepatic<br />

Pediatric Use<br />

Pregnancy Can Drug Be Crushed<br />

Dosing Dosing<br />

Category<br />

Ganciclovir Yes No The safety and efficacy of gancicloivr<br />

in pediatric patients have not been<br />

established. Use of ganciclovir in this<br />

patient population warrants extreme<br />

caution due to the probability of<br />

long-term carcinogenicity and<br />

reproductive toxicity. Administration<br />

should be undertaken only after<br />

careful evaluation and only if the<br />

potential benefits of treatment<br />

outweigh the risks.<br />

C According to the package insert,<br />

ganciclovir capsules should not be<br />

opened or crushed. The<br />

manufacturer reports no data are<br />

available regarding opening<br />

capsules or use as an oral<br />

suspension.<br />

Valganciclovir Yes No Safety and efficacy in pediatric<br />

patients have not been established.<br />

Acyclovir Yes No Safety and efficacy of oral<br />

formulations of acyclovir in pediatric<br />

patients under 2 years of age have not<br />

been established. The intravenous<br />

formulation is FDA-approved for the<br />

treatment of neonatal HSV infection;<br />

dosing recommendations are<br />

available.<br />

Valacyclovir Yes No Safety and efficacy in prepubertal<br />

pediatric patients have not been<br />

established.<br />

Famciclovir Yes No Safety and efficacy in children under<br />

the age of 18 years have not been<br />

established.<br />

Cidofovir Yes No Safety and efficacy in children have<br />

not been established. Use of cidofovir<br />

C<br />

B<br />

B<br />

B<br />

C<br />

According to the package insert,<br />

valganciclovir tablets should not be<br />

broken or crushed. However,<br />

information is available from the<br />

manufacturer regarding the<br />

preparation of an extemporaneous<br />

oral liquid prepared from<br />

valganciclovir tablets.<br />

The manufacturer reports no data<br />

are available regarding opening<br />

capsules or crushing tablets as an<br />

oral suspension formulation is<br />

available.<br />

The manufacturer reports that<br />

caplets are scored and can be<br />

broken. The pharmacokinetics of<br />

crushed tablets have not been<br />

specifically evaluated. However,<br />

information on the preparation of<br />

an extemporaneous suspension<br />

from 500 mg tablets is available<br />

from the manufacturer and has<br />

been evaluated for formulation,<br />

stability, sterility, and palatability.<br />

The manufacturer reports that there<br />

is no information regarding<br />

splitting or crushing famciclovir<br />

tablets.<br />

Intravenous solution is only<br />

formulation currently available.<br />

241


in children with AIDS warrants<br />

extreme caution due to the risk of<br />

long-term carcinogenicity and<br />

reproductive toxicity. Administration<br />

should only be undertaken after<br />

careful evaluation and only if the<br />

potential benefits of treatment<br />

outweigh the risks.<br />

Ribavirin Yes* No Ribavirin capsules and oral solution<br />

are indicated in patients 3 years of age<br />

and older. Importantly, however,<br />

suicidal ideation or attempts occurred<br />

more frequently among pediatric<br />

patients, primarily adolescents,<br />

compared to adult patients during<br />

treatment and off-therapy follow-up.<br />

Safety and efficacy of ribavirin tablets<br />

(ie, Copegus ® ) in pediatric patients<br />

under the age of 18 years have not<br />

been established.<br />

Adefovir Yes No Safety and efficacy in pediatric<br />

patients have not been established.<br />

*Ribavirin should not be used in patients with a creatinine clearance less than 50 mL/min.<br />

X<br />

C<br />

According to the manufacturers, no<br />

information is available regarding<br />

crushing of ribavirin tablets (ie,<br />

Copegus ® ) or opening of ribavirin<br />

capsules (ie, Rebetol ® ). An oral<br />

solution formulation is available.<br />

According to the manufacturer,<br />

there is no data available regarding<br />

crushing or splitting adefovir<br />

tablets.<br />

242


VIII.<br />

Comparative Effectiveness of the Antiviral Nucleosides and Nucleotides<br />

Table 8. Outcomes Evidence for the Antiherpetic Agents<br />

Study Sample<br />

Size<br />

Treatment/<br />

Duration<br />

Shafran<br />

SD, et al 29 n=559 Famciclovir 750mg<br />

QD versus famciclovir<br />

500mg BID versus<br />

famciclovir 250mg<br />

TID versus acyclovir<br />

800mg 5 times daily<br />

for 7 days<br />

Warkentin<br />

DI, et al 30 n=151 Valacyclovir 500mg<br />

BID versus<br />

valacyclovir 250mg<br />

BID versus acyclovir<br />

400mg TID<br />

Shen MC, n=55 Famciclovir 250mg<br />

et al 31 TID versus acyclovir<br />

800mg 5 times daily<br />

Lin WR,<br />

et al 32 n=57 Valacyclovir 1000mg<br />

TID versus acyclovir<br />

800mg 5 times daily<br />

for 7 days<br />

Tyring S,<br />

et al 33 n=148 Famciclovir 500mg<br />

TID versus acyclovir<br />

800mg 5 times daily<br />

for 10 days<br />

243<br />

Results<br />

In comparing various doses of famciclovir to acyclovir for the treatment of herpes<br />

zoster in immunocompetent adults:<br />

• No significant differences between treatment groups with respect to time to full<br />

crusting, loss of vesicles/ulcers/crusts, cessation of new lesion formation, 50%<br />

reduction in area of affected skin, and resolution of acute pain.<br />

• There were no significant differences in the frequency of adverse events between<br />

treatment groups.<br />

• Agents had comparable efficacy in healing cutaneous herpes zoster.<br />

In comparing valacyclovir 500 mg BID (n=48), valacyclovir 250mg BID (n=52), and<br />

acyclovir 400mg TID (n=51) for HSV prophylaxis in neurtopenic patients, where<br />

therapies were given for the duration of neutropenia:<br />

• The absence of HSV infection was similar among the treatment groups:<br />

acyclovir 96%, valacyclovir 500mg 95%, valacyclovir 250mg 100% (P=0.08).<br />

• The overall rates of adverse events were similar in the 3 treatment groups.<br />

In comparing famcilovir (n=27) to acyclovir (n=28) for the treatment of acute,<br />

uncomplicated herpes zoster in immunocompetent adults:<br />

• Treatment was initiated within 72 hours of onset of the zoster rash and was<br />

continued for 7 days.<br />

• Famciclovir and acyclovir had comparable efficacy with respect to healing<br />

cutaneous lesions (indicated by time to full crusting), resolution of acute pain,<br />

loss of vesicles, and loss of crusts.<br />

• Constipation, hematuria, and glycosuria were the most commonly reported<br />

adverse events. Famciclovir had a more favorable adverse event profile than<br />

acyclovir.<br />

In comparing valacyclovir (n=32) to acyclovir (n=25) for the treatment of acute,<br />

uncomplicated herpes zoster:<br />

• Patients presenting with herpes zoster within 72 hours after onset of zoster rash<br />

were included.<br />

• Valacyclovir significantly shortened the time necessary for resolution of herpes<br />

zoster associated pain compared to acyclovir.<br />

• There were no significant differences between groups with respect to frequency<br />

or severity of adverse events.<br />

• The authors concluded that valacyclovir accelerates the resolution of herpes<br />

zoster pain with a simpler dosing schedule, while maintaining a similar safety<br />

profile as acyclovir.<br />

In comparing famciclovir to acyclovir for the treatment of herpes zoster in<br />

immunocompromised patients 12 years of age or older:<br />

• Famciclovir and acyclovir were comparable with respect to the number of<br />

patients reporting new lesion formation while on therapy (77% versus 73%,<br />

respectively).<br />

• There were no differences between treatment regimens in time to cessation of<br />

new lesion formation, full crusting, complete healing of lesions, or resolution of<br />

acute pain.<br />

• The adverse event profile was comparable between treatment groups.<br />

Chosidow<br />

O, et al 34 n=204 Famciclovir 125mg<br />

BID versus acyclovir<br />

In comparing famciclovir to acyclovir for the treatment of recurrent genital herpes<br />

infection:<br />

200mg 5 times daily • Mean healing time was 5.1 days with famciclovir and 5.4 days with acyclovir<br />

for 5 days<br />

(P=0.82).<br />

• No difference was observed in the proportion of patients having complete<br />

healing on different days of evaluation. Additionally, there was no difference in<br />

the duration until the complete resolution of all symptoms.<br />

• The adverse event profile was comparable between treatment groups.<br />

• Famciclovir and acyclovir had comparable safety and efficacy in the treatment of<br />

recurrent genital herpes.<br />

Romanow n=293 Famciclovir 500mg In comparing famcilovir to acyclovir for the treatment of mucocutaneous HSV


ski B, et<br />

al 35<br />

BID versus acyclovir<br />

400mg 5 times daily<br />

for 7 days<br />

Tyring<br />

SK, et al 36 n=1,250 Valacyclovir 1000mg<br />

BID versus acyclovir<br />

200mg 5 times daily<br />

versus placebo for<br />

5 days<br />

Fife KH,<br />

et al 37 n=643 Valacyclovir 1000mg<br />

BID versus acyclovir<br />

200mg 5 times daily<br />

for 10 days<br />

Bodswort<br />

h NJ, et<br />

al 38 n=739 Valacyclovir 500mg<br />

BID versus acyclovir<br />

200mg 5 times daily<br />

for 5 days<br />

Tyring<br />

SK, et al 39 n=597 Valacyclovir 1000mg<br />

TID versus<br />

famciclovir 500mg<br />

TID for 7 days<br />

infection in HIV-infected patients:<br />

• Patients initiated therapy within 48 hours of the first appearance of lesions.<br />

• Famciclovir and acyclovir were comparable with respect to prevention of new<br />

lesion formation (new lesions in 16.7% and 13.3% of patients, respectively).<br />

• Famciclovir and acyclovir treatment groups were also comparable in time to<br />

complete healing (median 7 days in each group), cessation of viral shedding<br />

(median of 2 days), and loss of lesion-associated symptoms (median 4 days).<br />

• The incidence of adverse events was comparable between treatment groups.<br />

• Famciclovir and acyclovir are equally safe and effective for the treatment of<br />

mucocutaneous infections in HIV-infected patients; however, famciclovir has a<br />

more convenient dosing schedule.<br />

In comparing valacyclovir, acyclovir, and placebo for the treatment of recurrent<br />

genital herpes infections:<br />

• Valacyclovir and acyclovir were significantly more effective than placebo in<br />

accelerating resolution of herpes episodes, time to lesion healing, and resolution<br />

of pain.<br />

• Valacyclovir and acyclovir demonstrated comparable efficacy with respect to the<br />

above parameters.<br />

• The incidence of adverse events did not differ among the 3 treatment groups.<br />

In comparing valacyclovir to acyclovir for the treatment of adults with a first episode<br />

of genital herpes:<br />

• Valacyclovir and acyclovir were comparable with respect to duration of viral<br />

shedding, time to healing, duration of pain, and time to resolution of all<br />

symptoms.<br />

• Adverse events were comparable between treatment groups. Overall, adverse<br />

events were infrequent and mild in severity.<br />

In comparing valacyclovir to acyclovir for the treatment of immunocompetent adults<br />

with recurrent genital herpes infections:<br />

• Valacyclovir and acyclovir were comparable with respect to duration of signs<br />

and symptoms (including lesion healing and pain).<br />

• Cessation of viral shedding was rapid and comparable between treatment groups.<br />

• Adverse events were infrequent and generally mild in severity. Incidence of<br />

adverse events was similar between treatment groups.<br />

In comparing valacyclovir to famciclovir for the treatment of immunocompetent<br />

adults, ages 50 years and older, with herpes zoster:<br />

• Patients were required to present within 72 hours of rash onset for inclusion.<br />

• The valacyclovir and famciclovir groups were comparable with respect to<br />

resolution of zoster-associated pain, rash healing, and analyses of postherpetic<br />

neuralgia.<br />

• Safety profiles of valacyclovir and famciclovir were similar; headache and<br />

nausea were the most common adverse events.<br />

• Valacyclovir was comparable to famciclovir treatment in accelerating the<br />

resolution of zoster-associated pain and postherpetic neuralgia.<br />

244


Table 9. Outcomes Evidence for the Treatment or Prevention of Cytomegalovirus<br />

Study Sample<br />

Size<br />

Treatment/<br />

Duration<br />

Results<br />

Paya C, et<br />

al. 40 n=364 Valganciclovir 900mg<br />

QD versus oral<br />

ganciclovir 1000mg<br />

TID<br />

Szer, et<br />

al. 41 n=58 Oral ganciclovir<br />

3000mg QD versus IV<br />

ganciclovir 5mg/kg<br />

three times weekly<br />

Martin<br />

DF, et al 42 n=160 Oral valganciclovir<br />

900mg BID for<br />

3 weeks, followed by<br />

900mg QD for 1 week<br />

versus IV ganciclovir<br />

5mg/kg BID for<br />

3 weeks, followed by<br />

5mg/kg QD for<br />

1 week<br />

Winston<br />

DJ, et al 43 n=64 Sequential IV and oral<br />

ganciclovir (6mg/kg<br />

IV QD for 2 weeks,<br />

followed by 1000mg<br />

every 8 hours oral<br />

ganciclovir) versus<br />

prolonged IV<br />

ganciclovir (6mg/kg<br />

QD for 2 weeks,<br />

followed by 6mg/kg<br />

QD 5 days per week)<br />

Jabs DA 44 n=61 Ganciclovir<br />

ophthalmic implant<br />

plus oral ganciclovir<br />

1000mg TID versus<br />

IV cidofovir 5mg/kg<br />

weekly for 2 doses,<br />

followed by 5mg/kg<br />

every other week<br />

In comparing valganciclovir and oral ganciclovir for the prophylaxis of CMV in solid<br />

organ transplant patients who were CMV-seronegative but receiving organs from<br />

CMV-seropositive donors:<br />

• At 6 months, CMV disease had developed in 12.1% and 15.2% of valganciclovir<br />

and ganciclovir patients, respectively.<br />

• At 12 months, CMV disease had developed in 17.2% and 18.4% of<br />

valganciclovir and ganciclovir patients, respectively. Additionally, the incidence<br />

of investigator-treated CMV disease events was comparable between treatment<br />

groups.<br />

• Oral valganciclovir was as effective and well-tolerated as oral ganciclovir for<br />

CMV prevention in high-risk solid organ transplant patients.<br />

In comparing oral ganciclovir to IV ganciclovir in patients at risk for CMV disease<br />

following allogenic BMT:<br />

• CMV-seropositive patients or patients receiving bone marrow from<br />

CMV-seropositive donors were randomized to IV ganciclovir (n=27) or oral<br />

ganciclovir (n=31) from engraftment to day 84.<br />

• Renal dysfunction, transfusion requirements, and significant nausea and<br />

vomiting were similar between treatment groups.<br />

• There were no documented cases of CMV disease in either treatment group<br />

during the study.<br />

• Oral ganciclovir was determined to be an effective, well-tolerated alternative to<br />

IV ganciclovir for prophylaxis of CMV disease.<br />

In comparing oral valganciclovir (n=80) to IV ganciclovir (n=80) for induction<br />

therapy for newly diagnosed CMV retinitis in patients with AIDS:<br />

• Progression of CMV was observed in 10% of patients receiving IV ganciclovir<br />

and 9.9% of patients receiving oral valganciclovir.<br />

• 77% of patients receiving IV ganciclovir and 71.9% receiving oral valganciclovir<br />

had a satisfactory response to induction therapy.<br />

• Median time to progression of retinitis was 125 days for IV ganciclovir and<br />

160 days for oral valganciclovir.<br />

• Adverse events were similar between treatment groups.<br />

• Oral valganciclovir was as effective as IV ganciclovir for induction treatment<br />

and long-term management of CMV retinitis in patients with AIDS.<br />

In comparing sequential IV and oral ganciclovir (n=32) to prolonged IV ganciclovir<br />

(n=32) for the prevention of CMV disease in CMV-seronegative liver transplant<br />

recipients with CMV-positive donors:<br />

• CMV disease occurred in 9.3% of patients receiving oral ganciclovir and 12.5%<br />

receiving IV ganciclovir within the first year after transplantation (P>0.2).<br />

• There were no deaths from CMV in either study group.<br />

• Both oral and IV ganciclovir were generally well-tolerated.<br />

• Results indicated that oral ganciclovir is as effective as IV ganciclovir for<br />

maintenance therapy after induction with IV ganciclovir.<br />

In comparing the ganciclovir ophthalmic implant plus oral ganciclovir to intravenous<br />

cidofovir for the treatment of cytomegalovirus retinitis in patients with AIDS:<br />

• Mortality was similar between treatment groups: 0.41 per person-year in<br />

ganciclovir patients and 0.49 per person-year in cidofovir patients (P=0.59).<br />

• Ocular outcomes (e.g., retinitis progression, loss of visual acuity) were similar<br />

between treatment groups.<br />

• Vitreous hemorrhage was more common in ganciclovir-treated patients (0.13 per<br />

person-year) than in cidofovir-treated patients (no cases) (P=0.14).<br />

• Uveitis was more common in the cidofovir group (0.35 per person-year)<br />

compared to the ganciclovir group (0.09 per person-year) (P=0.066).<br />

• Nephrotoxicity (serum creatinine greater than 1.6mg/dL) occurred at a rate of<br />

0.18 per person-year in the ganciclovir group and 0.48 per person-year in the<br />

cidofovir group (P=0.10).<br />

• Although the sample size for this study was small, the results suggest that the<br />

use of a ganciclovir implant plus oral ganciclovir and the use of intravenous<br />

245


Lalezari<br />

JP, et al. 45 n=48 Cidofovir 5mg/kg<br />

once weekly for<br />

2 weeks, followed by<br />

once every other week<br />

Lalezari<br />

JP, et al 46 n=150 Cidofovir 5mg/kg<br />

once weekly for<br />

2 weeks, followed by<br />

5mg/kg or 3mg/kg<br />

once every other week<br />

Ocular<br />

Complicat<br />

ions of<br />

AIDS<br />

Research<br />

Group 47 n=64 Cidofovir 5mg/kg<br />

once weekly for<br />

2 weeks, followed by<br />

3mg/kg once every<br />

2 weeks versus<br />

cidofovir 5mg/kg once<br />

weekly for 2 weeks,<br />

followed by 5mg/kg<br />

once weekly every<br />

2 weeks versus<br />

deferral of therapy<br />

until progression of<br />

retinitis<br />

cidofovir are similar for controlling CMV retinitis but have different adverse<br />

event profiles.<br />

In comparing immediate (n=25) with deferred (n=23) cidofovir treatment in patients<br />

with AIDS and previously untreated CMV retinitis:<br />

• Median time to progression of CMV retinitis in the deferred group was 22 days<br />

compared to 120 days in the immediate treatment group (P


Perillo R,<br />

et al 50 n=5 Adefovir 5-30mg QD In 5 patients with chronic HBV who developed resistance to lamivudine after 9-19<br />

months of therapy:<br />

• 4 of the patients included developed resistance after liver transplantation and 1<br />

patient had stable cirrhosis.<br />

• 2-4 log 10 reductions in serum HBV DNA levels were observed in 4 patients after<br />

receiving adefovir therapy. The fifth patient became negative for serum HBV<br />

DNA after retransplantation in conjunction with hepatitis B immunoglobulin.<br />

• Serum alanine transaminase levels occurred in 4 patients.<br />

• The authors concluded that adefovir can be effective in treating<br />

lamivudine-resistant mutants of HBV.<br />

Schiff ER,<br />

et al 51 n=324 Adefovir 10mg QD In evaluating the effects of adefovir in pre- and post-liver transplant patients with<br />

recurrent chronic HBV and evidence of lamivudine-resistance:<br />

• 81% of pre-transplant patients and 34% of post-transplant patients who received<br />

48 weeks of treatment achieved undetectable serum HBV DNA levels.<br />

• Serum alanine aminotransferase, albumin, bilirubin, and prothrombin time<br />

normalized in 76%, 81% 50%, and 83% of pre-transplant patients and 49%,<br />

76%, 75%, and 20% of post-transplant patients, respectively.<br />

• Genotype revealed that 98% of 122 HBV baseline samples were lamivudine<br />

resistant.<br />

• No adefovir resistance mutations were identified after 48 weeks.<br />

• Adverse events in patients receiving adefovir were generally mild to moderate in<br />

severity.<br />

Benini F,<br />

et al 52 n=43 Ribavirin 1000mg or<br />

1200mg QD plus<br />

interferon-alfa 2b<br />

In evaluating the combination of ribavirin and interferon alfa-2b for the treatment of<br />

chronic HCV infection in patients experiencing relapses or non-responders to prior<br />

high-dose interferon:<br />

3MU 3 times per • Normalization of serum alanine aminotransferase levels and undetectable HCV<br />

week for 24 or 48 RNA were seen in 58.1% and 30.2% of patients, respectively.<br />

weeks • No significant difference in response was identified for previous non-responders<br />

of interferon therapy versus patients with relapses.<br />

• At the end of followup, 3 (7%) patients had a sustained response (2 interferon<br />

non-responders, 1 relapser).<br />

Brau N, et<br />

al 53 n=107 Interferon alfa-2b<br />

3MU 3 times per<br />

week with placebo for<br />

In evaluating the combination of ribavirin and interferon alfa-2b (n=53) versus<br />

interferon alfa-2b with placebo for 16 weeks followed by ribavirin 800 mg QD (n=54)<br />

for the treatment of chronic HCV infection in patients coinfected with HIV:<br />

16 weeks, followed by • Sustained viral response rate (ie, undetectable HCV RNA at 24 weeks<br />

ribavirin 800mg QD posttherapy) was comparable between groups: immediated ribavirin 11.3%,<br />

versus interferon delayed ribavirin 5.6% (P=0.32).<br />

alfa-2b (same dose) • At week 12, CD4 cell count increased in the immediate-ribavirin group (+4.1%,<br />

plus ribavirin 800mg P


Perillo R,<br />

et al 55 n=412 Interferon alfa-2b<br />

(3MU 3 times weekly)<br />

plus ribavirin<br />

1000-1200 mg QD<br />

versus peginterferon<br />

monotherapy (180mcg<br />

once weekly)<br />

Manns<br />

MP, et<br />

al 56 n=1530 Interferon alfa-2b<br />

(3 MU 3 times<br />

weekly) plus ribavirin<br />

1000-1200 mg QD<br />

versus peginterferon<br />

alfa-2b (1.5mcg/kg<br />

each week) plus<br />

ribavirin 800 mg QD<br />

versus peginterferon<br />

alfa-2b (1.5mcg/kg<br />

each week for<br />

4 weeks, then<br />

0.5mcg/kg each week)<br />

plus ribavirin<br />

1000-1200 mg QD for<br />

48 weeks<br />

McHutchi n=912 Interferon alfa-2b<br />

et al 57 interferon alfa-2b<br />

nson JG,<br />

(standard dose) versus<br />

(standard dose) plus<br />

ribavirin<br />

1000-1200 mg QD for<br />

24 or 48 weeks<br />

In evaluating the impact of peginterferon monotherapy versus combined interferon<br />

alfa-2b/ribavirin therapy on quality of life, work productivity, and medical resource<br />

utilization:<br />

• Patients receiving peginterferon monotherapy reported less impairment with<br />

respect to all measures of work functioning and productivity compared to<br />

patients receiving interferon alfa-2b/ribavirin.<br />

• The authors concluded that patients receiving peginterferon had improved work<br />

productivity, less activity impairment, better adherence, and decreased need for<br />

prescription drugs to treat adverse events.<br />

In assessing peginterferon alfa-2b with ribavirin versus interferon alfa-2b plus<br />

ribavirin for the treatment of chronic HCV:<br />

• Sustained virologic response was significantly higher in the higher-dose<br />

peginterferon group (54%) than the lower-dose peginterferon group (47%) or<br />

interferon group (47%).<br />

• In patients with genotype 1 HCV, sustained virologic response was 42%, 34%,<br />

and 33%, in the high-dose peginterferon, low-dose peginterferon, and interferon<br />

groups, respectively.<br />

• The rate of sustained virologic response for patients with genotypes 2 or 3 was<br />

about 80% for all treatment groups.<br />

• The authors concluded that peginterferon 1.5mcg/kg per week with ribavirin was<br />

the most effective therapy.<br />

In assessing the efficacy and safety of interferon alfa-2b monotherapy compared to a<br />

combination of interferon alfa-2b plus ribavirin for the initial therapy of patients with<br />

chronic HCV infection:<br />

• Sustained virologic response was higher in patients on combination therapy at<br />

both 24 and 48 weeks versus patients on interferon monotherapy.<br />

• In patients with genotype 1 HCV, the best response was observed in patients<br />

receiving combination interferon/ribavirin therapy.<br />

• Histologic improvement was also more common in patients on combination<br />

therapy versus monotherapy.<br />

• Drug dosages had to be reduced more frequently in the combination group and<br />

more treatment discontinuations occurred in these patients.<br />

• The authors concluded that combination therapy with interferon and ribavirin<br />

was more effective than treatment with interferon alone.<br />

Table 11. Additional Outcomes Evidence for Aerosolized Ribavirin in the Treatment of Respiratory Syncytial Virus<br />

Study Sample<br />

Size<br />

Treatment/<br />

Duration<br />

Results<br />

Guergueri n=41 Ribavirin 6g/300mL<br />

al 58<br />

an AM, et<br />

versus placebo<br />

Rodriguez n=42 Aerosolized ribavirin<br />

WJ, et al 59 versus placebo<br />

In assessing the effect of aerosolized ribavirin on previously healthy infants requiring<br />

mechanical ventilation for respiratory distress secondary to RSV:<br />

• There were no significant differences between ribavirin and placebo with respect to<br />

ventilation, length of oxygen therapy, length of stay in intensive care, and total<br />

duration of hospital stay.<br />

• Both ribavirin and normal saline placebo were well-tolerated and no deaths<br />

occurred during the study.<br />

• Ribavirin was not effective in producing significant clinical outcomes in patients<br />

with RSV.<br />

In a prospective follow-up study of children who were previously healthy, were<br />

premature, or who had chronic pulmonary disease:<br />

• More reactive airway disease, wheezing, and pneumonia was observed in the<br />

placebo group (n=12) than the ribavirin group (n=23): mean score 22.3 and 15.8,<br />

respectively (P=0.07). For all years of the study the mean score was 22 for the<br />

placebo group (n=11) and 16 for the ribavirin group (n=22) (P=0.10).<br />

• Seven out of 13 ribavirin patients had normal or mild pulmonary function test<br />

results versus zero of 6 placebo patients (P=0.04).<br />

• Weighted severity scores suggested a long-term beneficial effect of ribavirin;<br />

248


Jefferson<br />

LS, et al 60 n=13 Ribavirin 6g/300mL<br />

over 18 hr/day versus<br />

ribavirin 6g/100mL<br />

over 2 hr divided TID<br />

Edell D, n=45 Ribavirin 6g/100mL<br />

et al 61 over 2 hr divided TID<br />

versus conservative<br />

treatment<br />

Moler n=223 Ribavirin versus no<br />

al 62<br />

FW, et<br />

therapy<br />

Long CE, n=54 Ribavirin versus<br />

et al 63 placebo<br />

however, the authors stated that larger numbers should be evaluated.<br />

In a study to evaluate the effect of ribavirin on respiratory system mechanics in pediatric<br />

patients with suspected RSV requiring mechanical ventilation:<br />

• Authors concluded that aerosolized ribavirin in infants and young children does not<br />

worsen respiratory system mechanics.<br />

In assessing the effect of aerosolized ribavirin compared to conservative treatment for<br />

previously healthy infants with severe RSV:<br />

• Conservative treatment included oxygen, IV fluids, albuterol nebulizers, IV<br />

corticosteroids, and oral ranitidine.<br />

• During a prospective 1-year follow-up period, the ribavirin group had significantly<br />

fewer episodes and reduced severity of reactive airway disease compared to the<br />

group receiving conservative therapy.<br />

• The ribavirin group also had significantly fewer hospitalizations related to<br />

respiratory illness compared to conservative therapy (25 versus 90 days per<br />

100 patients per year, respectively).<br />

• The authors concluded that early ribavirin therapy for RSV reduced the incidence<br />

and severity of reactive airway disease and decreased hospitalizations related to<br />

respiratory illness.<br />

In assessing the effect of aerosolized ribavirin compared to no therapy in previously<br />

healthy infants with RSV-associated respiratory failure on mechanical ventilation:<br />

• Prospective cohort study.<br />

• Use of ribavirin (n=91) was associated with prolonged duration of mechanical<br />

ventilation (P


IX.<br />

Conclusions<br />

The efficacy of acyclovir, famciclovir, and valacyclovir against HSV and VZV infections were<br />

similar based on several comparative studies. The CDC guidelines suggest all three antiherpetic<br />

virus agents can be used for the treatment and suppression of genital herpes. The difference in<br />

these three agents is the dosing schedule, with newer agents offering more convenient dosing.<br />

Selection of one agent for the preferred drug list should provide sufficient coverage for patients<br />

with HSV or VZV infections. Acyclovir is the only agent with an available generic product and<br />

the only agent recommended by the AAP for treatment of chickenpox in patients at risk for more<br />

severe disease. Treatment with these agents would be considered applicable to general use in the<br />

population.<br />

The efficacy of oral ganciclovir, intravenous ganciclovir, and oral valganciclovir for the treatment<br />

of CMV retinitis were similar based on several comparative studies. However, oral ganciclovir is<br />

indicated only for maintenance therapy of CMV retinitis in patients with stable retinitis following<br />

appropriate induction therapy with intravenous ganciclovir. Ganciclovir capsules are the only<br />

generic formulation available. Cidofovir is also an effective treatment option for CMV retinitis;<br />

however, comparative data for cidofovir versus ganciclovir or valganciclovir are lacking.<br />

Nephrotoxicity, which may be irreversible, is the major adverse event limiting duration of<br />

cidofovir treatment in a significant number of patients. Because there are only a limited number<br />

of medications available for the treatment of CMV retinitis, cidofovir represents a useful<br />

alternative in patients who do not respond to ganciclovir or valganciclovir therapy. The treatments<br />

for CMV retinitis do not represent therapeutic agents applicable to general use of the population.<br />

Oral ribavirin is recommended in combination with peginterferon for the treatment of chronic<br />

HCV infection by the American Association for the Study of Liver Diseases. Rebetol ® is<br />

available in two formulations (capsules and oral solution) and is indicated in combination with<br />

either interferon alfa-2b or peginterferon alfa-2b. Copegus ® is available as a tablet and is only<br />

indicated in combination with peginterferon alfa-2a. Treatment of HCV is not applicable to<br />

general use of the population. Use of ribavirin also is dependent on the brand of interferon used as<br />

combination products are available.<br />

Adefovir, lamivudine, and interferon-alfa are all recommended as options for first-line therapy of<br />

chronic HBV infection. 5,6 Adefovir is also recommended for use in patients that have<br />

demonstrated lamivudine resistance. Lamivudine and interferon have been evaluated in other<br />

reviews; therefore, comparative safety and efficacy data have not been extensively covered in this<br />

document. However, due to the relatively few options for therapy of chronic HBV and the utility<br />

of adefovir in the face of lamivudine-resistance, adefovir represents a valuable therapeutic option.<br />

Use of these agents for HBV is not applicable to use in the general population.<br />

Although aerosolized ribavirin demonstrates good in vitro activity against RSV, clinical efficacy<br />

data have been conflicting. Due to the lack of definitive efficacy, and potential toxicities of this<br />

medication, ribavirin is not generally used for the treatment of RSV, and would not be considered<br />

general use within the population.<br />

Within this class, the antiherpetic agents are important for general use in the population. All brand<br />

antiherpetic agents in this class are comparable to each other and to the generics and OTC<br />

products in the class and offer no significant clinical advantage over other alternatives in general<br />

use. Additionally, the treatments for CMV, HCV, HBV, and RSV are not within the scope of<br />

general use in the population, and should be available for their indicated special<br />

needs/circumstances via medical justification through the prior authorization process.<br />

X. Recommendations<br />

No brand antiviral nucleoside/ nucleotide is recommended for preferred status.<br />

250


References<br />

1. Corey LC. Herpes Simplex Virus. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and<br />

Practice of Infectious Diseases. 5 th ed. Philadelphia, Pa: Churchill Livingstone; 2000:1564-1575.<br />

2. Knodel LC. Sexually Transmitted Disease. In: Dipiro JT, Talbert RL, Yee GC,<br />

Matzke GR, Wells BG, Posey LM, eds. <strong>Pharmacotherapy</strong>: A Pathophysiologic Approach. 5 th ed.<br />

New York, NY: McGraw-Hill; 2002:1997-2016.<br />

3. Whitley RJ. Varicella-Zoster Virus. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and<br />

Practice of Infectious Diseases. 5 th ed. Philadelphia, Pa: Churchill Livingstone; 2000:1580-1585.<br />

4. Crumpacker CS. Cytomegalovirus. In: Mandell GL, Bennett JE, Dolin R, eds.<br />

Principles and Practice of Infectious Diseases. 5 th ed. Philadelphia, Pa: Churchill Livingstone;<br />

2000:1586-1596.<br />

5. Lok ASF, McMahon BJ. American Association for the Study of Liver Diseases<br />

(AASLD) Practice Guidelines for Chronic Hepatitis B. December, 2003. Available at:<br />

www.aasld.org. Accessed September 20, 2004.<br />

6. Lok ASF, McMahon BJ. American Association for the Study of Liver Diseases<br />

(AASLD) practice guideline for chronic hepatitis B: update of recommendations. Hepatology.<br />

2004;1-5. Available at: www.aasld.org. Accessed September 20, 2004.<br />

7. Strader DB, Wright T, Thomas DL, Seeff LB. American Association for the Study of Liver<br />

Diseases (AASLD) practice guideline for diagnosis, management, and treatment of hepatitis C.<br />

Hepatology. 2004;1147-1171. Available at www.aasld.org. Accessed September 20, 2004.<br />

8. National Institutes of Health. Consensus Development Conference Statement on the Management<br />

of Hepatitis C: 2002. National Institutes of Health. Available at: http://consensus.nih.gov.<br />

Accessed September 20, 2004.<br />

9. Polak MJ. Respiratory syncytial virus (RSV): overview, treatment, and prevention strategies.<br />

NBIN. 2004;14(1):15-23.<br />

10. National Center for HIV, STD, and TB Prevention, Division of Sexually Transmitted Diseases.<br />

Sexually Transmitted Treatment Guidelines 2002. Centers for Disease Control and Prevention<br />

(CDC). Available at www.cdc.gov/mmwr. Accessed September 20, 2004.<br />

11. American Academy of Pediatrics (AAP). In: Pickering LK, ed. 2003 Red Book: Report of the<br />

Committee on Infectious Diseases. 26 th ed. Elk Grove Village, Ill: American Academy of<br />

Pediatrics;2003:672-686.<br />

12. Gnann JW Jr, Whitley RJ. Herpes zoster [clinical practice]. N Engl J Med. 2002;347(5):340-346.<br />

13. Zovirax [package inserts]. Research Triangle Park, NC: GlaxoSmithKline; 2003.<br />

14. Valtrex [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2003.<br />

15. Famvir [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2002.<br />

16. US Public Health Service and the Infectious Disease Society of America. Guidelines for<br />

Preventing Opportunistic Infections Among HIV Infected Persons 2002. Centers for Disease<br />

Control and Infection. Available at: www.cdc.gov/mmwr. Accessed September 20, 2004.<br />

17. Cytovene-IV, Cytovene [package insert]. Nutley, NJ: Roche Laboratories Inc; 2000.<br />

18. Vistide [package insert]. Foster City, Calif: Gilead Sciences, Inc.; 2000.<br />

19. Centers for Disease Control and Prevention, Infectious Disease Society of America, and the<br />

American Society of Blood and Marrow Transplantation. Guidelines for Preventing Opportunistic<br />

Infections Among Hematopoietic Stem Cell Transplant Recipients 2000. Available at:<br />

www.cdc.gov/mmwr. Accessed September 20, 2004.<br />

20. Valcyte [package insert]. Nutley, NJ: Roche Laboratories Inc; 2003.<br />

21. American Academy of Pediatrics (AAP). In: Pickering LK, ed. 2003 Red Book: Report of the<br />

Committee on Infectious Diseases. 26 th ed. Elk Grove Village, Ill: American Academy of<br />

Pediatrics;2003:523-528.<br />

22. Drug Facts and Comparisons. Facts and Comparisons. St. Louis, Mo; 2004.<br />

23. American Hospital Formulary Service, AHFS Drug Information. American Society of Health-<br />

System Pharmacists. Bethesda, Md; 2004.<br />

24. Copegus [package insert]. Nutley, NJ: Roche Laboratories Inc; 2004.<br />

25. Rebetol [package insert]. Kenilworth, NJ: Schering Corporation; 2003.<br />

26. Virazole [package insert]. Costa Mesa, Calif: Valeant Pharmaceuticals International; 2004.<br />

27. Hepsera [package insert]. Foster City, Calif: Gilead Sciences, Inc.; 2004.<br />

251


28. Hansten PD, Horn JR, ed. Drug Interactions: Analysis and Management. Facts and Comparisons<br />

Publishing Group. St. Louis, Mo; 2004.<br />

29. Shafran SD, Tyring SK, Ashton R, et al. Once, twice, or three times daily famciclovir compared<br />

with aciclovir for the oral treatment of herpes zoster in immunocompetent adults: a randomized,<br />

multicenter, double-blind clinical trial. J Clin Virol. 2004;29:248-253.<br />

30. Warkentin DI, Epstein JB, Campbell LM, et al. Valacyclovir versus acyclovir for HSV<br />

prophylaxis in neutropenic patients. Ann Pharmacother. 2002;36:1525-1531.<br />

31. Shen MC, Lin HH, Lee SSJ, Chen YS, Chiang PC, Liu YC. Double-blind, randomized, acyclovircontrolled,<br />

parallel-group trial comparing the safety and efficacy of famciclovir and acyclovir in<br />

patients with uncomplicated herpes zoster. J Microbiol Immunol Infect. 2004;37(2):1684-1182.<br />

32. Lin WR, Lin HH, Lee SSJ, et al. Comparative study of the efficacy and safety of valaciclovir<br />

versus acyclovir in the treatment of herpes zoster. J Microbiol Immunol Infect. 2001;34(2):138-<br />

142.<br />

33. Tyring S, Belanger R, Bezwoda W, Ljungman P, Boon R, Saltzman RL. A randomized,<br />

double-blind trial of famciclovir versus acyclovir for the treatment of localized dermatomal herpes<br />

zoster in immunocompromised patients. Cancer Invest. 2001;19(1):13-22.<br />

34. Chosidow O, Drouault Y, Leconte-Veyriac F, et al. Famciclovir vs. aciclovir in immunocompetent<br />

patients with recurrent genital herpes infections: a parallel-groups, randomized, double-blind trial.<br />

Br J Dermatol. 2001;144(4):818-824.<br />

35. Romanowski B, Aoki FY, Martel AY, Lavender EA, Parsons JE, Saltzman RL. Efficacy and<br />

safety of famciclovir for treating mucocutaneous herpes simplex infection in HIV-infected<br />

individuals. AIDS. 2000;14(9):1211-1217.<br />

36. Tyring SK, Douglas JM Jr, Corey L, Spruance SL, Esmann J. A randomized, placebo-controlled<br />

comparison of oral valacyclovir and acyclovir in immunocompetent patients with recurrent genital<br />

herpes infections. Arch Dermatol. 1998;134(2):185-191.<br />

37. Fife KH, Barbarash RA, Rudolph T, et al. Valaciclovir versus acyclovir in the treatment of<br />

first-episode genital herpes infection: results of an international, multicenter, double-blind,<br />

randomized clinical trial. Sex Transm Dis. 1997;24(8):481-486.<br />

38. Bodsworth NJ, Crooks RJ, Borelli S, et al. Valaciclovir versus aciclovir in patient initiated<br />

treatment of recurrent genital herpes: a randomized, double-blind clinical trial. Genitourin Med.<br />

1997;73(2):110-116.<br />

39. Tyring SK, Beutner KR, Tucker BA, Anderson WC, Crooks RJ. Antiviral therapy for herpes<br />

zoster: randomized, controlled clinical trial of valacyclovir and famciclovir therapy in<br />

immunocompetent patients 50 years and older. Arch Fam Med. 2000;9(9):863-869.<br />

40. Paya C, Humar A, Dominguez E, et al. Efficacy and safety of valgancilovir vs. oral ganciclovir for<br />

prevention of cytomegalovirus disease in solid organ transplant recipients. Am J Transplant.<br />

2004;4(4):611-620.<br />

41. Szer J, Durrant S, Schwarer AP, et al. Oral versus intravenous ganciclovir for the prophylaxis of<br />

cytomegalovirus disease after allogenic bone marrow transplantation. Intern Med J.<br />

2004;34(3):98-101.<br />

42. Martin DF, Sierra-Madero J, Walmsley S, et al. A controlled trial of valganciclovir as induction<br />

therapy for cytomegalovirus retinitis. N Engl J Med. 2002;346(15):1119-1126.<br />

43. Winston DJ, Busuttil RW. Randomized controlled trial of sequential intravenous and oral<br />

ganciclovir versus prolonged intravenous ganciclovir for long-term prophylaxis of<br />

cytomegalovirus disease in high-risk cytomegalovirus-seronegative liver transplant recipients with<br />

cytomegalovirus-seropositive donors. Transplant. 2004;77(2):305-308.<br />

44. Jabs DA. The ganciclovir implant plus oral ganciclovir versus parenteral cidofovir for the<br />

treatment of cytomegalovirus retinitis in patients with acquired immunodeficiency syndrome: the<br />

ganciclovir cidofovir cytomegalovirus retinitis trial. Am J Ophthalmol. 2001;131(4):457-467.<br />

45. Lalezari JP, Stagg RJ, Kuppermann BD, et al. Intravenous cidofovir for peripheral<br />

cytomegalovirus retinitis in patients with AIDS: a randomized, controlled trial. Ann Intern Med.<br />

1997;126(4):257-263.<br />

46. Lalezari JP, Holland GN, Kramer F, et al. Randomized, controlled study of the safety and efficacy<br />

of intravenous cidofovir for the treatment of relapsing cytomegalovirus retinitis in patients with<br />

AIDS. J Acquir Immune Defic Syndr Hum Retrovirol. 1998;17(4):339-344.<br />

47. Studies of Ocular Complications of AIDS Research Group in collaboration with the AIDS Clinical<br />

Trials Group. Parenteral cidofovir for cytomegalovirus retinitis in patients with AIDS: the<br />

252


HPMPC peripheral cytomegalovirus retinitis trial: a randomized, controlled trial. Ann Intern Med.<br />

1997;126(4):264-274.<br />

48. Marcellin P, Chang TT, Lim SG, et al. Adefovir dipivoxil for the treatment of hepatitis B e<br />

antigen-positive chronic hepatitis B. New Engl J Med. 2003;348(9):808-816.<br />

49. Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, et al. Adefovir dipivoxil for the treatment of<br />

hepatitis B e antigen-negative chronic hepatitis B. New Engl J Med. 2003;348(9):800-807.<br />

50. Perrillo R, Schiff E, Yoshida E, et al. Adefovir dipivoxil for the treatment of lamivudine-resistant<br />

hepatitis B mutants. Hepatology. 2000;32(1):129-134.<br />

51. Schiff ER, Lai CL, Hadziyannis S, et al. Adefovir dipivoxil therapy for lamivudine-resistant<br />

hepatits B in pre- and post-liver transplantation patients. Hepatology. 2003;38(6):1419-1427.<br />

52. Benini F, Distefano L, Baisini O, Pigozzi MG, Lanzini A. Efficacy and tolerability of combination<br />

therapy with interferon-alfa plus ribavirin in patients with chronic hepatitis c virus infection: a<br />

single-center study in relapsers and nonresponders to previous treatment with high-dose<br />

interferon-alfa monotherapy. Curr Ther Res. 2003;64(3):140-150.<br />

53. Brau N, Rodriguez TM, Prokupek D, et al. Treatment of chronic hepatitis c in HIV/HCVcoinfection<br />

with interferon (alpha)-2b + full-course vs. 16-week delayed ribavirin. Hepatology.<br />

2004;39(4):989-998.<br />

54. Laguno M, Murillas J, Blanco JL, et al. Peginterferon alfa-2b plus ribavirin compared with<br />

interferon alfa-2b plus ribavirin for treatment of HIV/HCV coinfected patients. AIDS.<br />

2004;18(13):27-36.<br />

55. Perrillo R, Rothstein KD, Rubin R, et al. Comparison of quality of life, work productivity and<br />

medical resource utilization of peginterferon alpha 2a vs the combination of interferon alpha 2b<br />

plus ribavirin as initial treatment in patients with chronic hepatitis C. J Viral Hep. 2004;11:157-<br />

165.<br />

56. Manns MP, McHutchison JG, Gordon SC, et al. Peginterferon alfa-2b plus ribavirin compared<br />

with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomized<br />

trial. Lancet. 2002;358(9286):958-965.<br />

57. McHutchison JG, Gordon SC, Schiff ER, et al. Interferon alfa-2b alone or in combination with<br />

ribavirin as initial treatment for chronic hepatitis C. Hepatitis Interventional Therapy Group. New<br />

Engl J Med. 1998;339(21):1485-1492.<br />

58. Guerguerian AM, Gauthier M, Lebel MH, Farrell CA, Lacroix J. Ribavirin in ventilated<br />

respiratory syncytial virus bronchiolitis. Am J Respir Crit Care Med. 1999;160:829-834.<br />

59. Rodriguez WJ, Arrobio J, Fink R, Kim HW, Milburn C. Prospective follow-up and pulmonary<br />

functions form a placebo-controlled randomized trial of ribavirin therapy in respiratory syncytial<br />

virus bronchiolitis. Ribavirin Study Group. Arch Pediatr Adolesc Med. 1999;153(5):469-474.<br />

60. Jefferson LS, Coss BJA, Englund JA, Walding D, Stein F. Respiratory system mechanics in<br />

patients receiving aerosolized ribavirin during mechanical ventilation for suspected respiratory<br />

syncytial viral infection. Pediatr Pulmonol. 1999;28(2):117-124.<br />

61. Edell D, Khoshoo V, Ross G, Salter K. Early ribavirin treatment of bronchiolitis: effect on longterm<br />

respiratory morbidity. Chest. 2002;122(3):935-939.<br />

62. Moler FW, Steinhart CM, Ohmit SE, Stidham GL. Effectiveness of ribavirin in otherwise well<br />

infants with respiratory syncytial virus-associated respiratory failure. J Pediatr. 1996;128(3):422-<br />

428.<br />

63. Long CE, Voter KZ, Barker WH, Hall CB. Long term follow-up of children hospitalized with<br />

respiratory syncytial virus lower respiratory tract infection and randomly treated with ribavirin or<br />

placebo. Pediatr Infect Dis J. 1997;16(11):1023-1028.<br />

64. Grant DM, Mauskopf JA, Bell L, et al. Comparison of valacyclovir and acyclovir for the<br />

treatment of herpes zoster in immunocompetent patients over 50 years of age: a cost consequence<br />

model. <strong>Pharmacotherapy</strong> 1997 Mar-Apr;17(2):333-41.<br />

65. Huse DM, Schainbaum D, Kirsch, et al. Economic evaluation of famciclovir in reducing the<br />

duration of postherptic neuralgia. Am J Health Syst Pharm 1997 May 15;54(10):1180-4.<br />

253


I. Overview<br />

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of Misc. Antivirals-Foscarnet<br />

AHFS 081892<br />

October 27, 2004<br />

Foscarnet sodium (“foscarnet”), an intravenous antiviral agent, is an organic analog of inorganic<br />

pyrophosphate, and is structurally unrelated to other antiviral agents. Although foscarnet was first<br />

synthesized in the 1920s, almost 60 years later it was found to inhibit influenza virus RNA<br />

polymerase. This led to investigation of foscarnet for potential activity against herpes simplex<br />

virus types 1 and 2 and against cytomegalovirus.<br />

Foscarnet was approved by the FDA in September 1991 and is currently indicated for the<br />

treatment of cytomegalovirus (CMV) retinitis in patients with acquired immunodeficiency<br />

syndrome (AIDS). Safety and efficacy of foscarnet sodium have not been established for<br />

treatment of other CMV infections (e.g., pneumonitis, gastroenteritis), congenital or neonatal<br />

CMV disease, or non-immunocompromised individuals. Foscarnet sodium is also indicated for<br />

the treatment of acyclovir-resistant mucocutaneous herpes simplex virus (HSV) infections in<br />

immunocompromised patients. Safety and efficacy of foscarnet sodium have not been established<br />

for treatment of other HSV infections (e.g., retinitis, encephalitis), congenital or neonatal HSV<br />

disease, or HSV in non-immunocompromised individuals.<br />

Foscarnet is also being investigated for treatment of cytomegalovirus disease, herpes simplex, and<br />

varicella-zoster infection in HIV-infected patients. Unlike ganciclovir, foscarnet is not associated<br />

with major myelosuppressive toxicity and does not require phosphorylation before it becomes<br />

activated. 1,2<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drug included in this<br />

review.<br />

Table 1. Misc. Antivirals in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name<br />

Foscarnet* Injectable 24mg/ml Foscavir<br />

*No Generic Available.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Cytomegalovirus Retinitis<br />

Ocular complications of acquired immunodeficiency syndrome (AIDS) are common. In HIVinfected<br />

patients 10-20% can expect to lose their sight in one or both eyes due to CMV retinitis.<br />

Foscarnet sodium is used for the treatment of CMV retinitis in patients with AIDS. The World<br />

Health Organization (WHO) guidelines recommend ganciclovir as the drug of choice and<br />

foscarnet as the next line agent for treatment and maintenance therapy of CMV retinitis. Foscarnet<br />

can induce stabilization or improvement of ocular manifestations, but the drug is only suppressive<br />

against the virus. The retinitis will recur and/or progress following discontinuation of therapy and<br />

may progress even in continued therapy in patients with AIDS unless improvement in<br />

immunocompetence is achieved. Combination therapy with foscarnet sodium and ganciclovir is<br />

1, 2, 3<br />

indicated for patients who have relapsed after monotherapy with either drug.<br />

254


Prevention of Recurrence of Cytomegalovirus Disease<br />

Suppressive or maintenance therapy (secondary prophylaxis) is recommended for HIV-infected<br />

patients. The Prevention of Opportunistic Infections Working Group of the U.S. Public Health<br />

Service and the Infectious Disease Society of America (USPHS/IDSA) in adults and adolescents<br />

recommends IV ganciclovir or IV foscarnet as drugs of choice. The USPHS/IDSA recommends<br />

that secondary prophylaxis should be restarted in adults or adolescents if the CD4+T cell count<br />

decreases to less than 100-150/mm 3 .<br />

Mucocutaneous Herpes Simplex Virus Infections<br />

Foscarnet is used in the management of acyclovir-resistant mucocutaneous herpes simplex virus<br />

(HSV-1 and HSV-2) infections in immunocompromised patients. Foscarnet is used in the<br />

management of severe HSV infections in HIV patients when the HSV is known or suspected of<br />

being caused by acyclovir-resistant strains of the virus. Since acyclovir-resistant HSV are<br />

resistant to valacyclovir and most are also resistant to famciclovir, the US Center for Disease<br />

Control and Prevention (CDC) recommends foscarnet until clinical resolution is attained.<br />

Varicella-Zoster Infections<br />

Although Foscarnet use in Varicella-Zoster infections is a non-FDA-approved indication,<br />

foscarnet has been used in the management of acyclovir-resistant Varicella-Zoster infections in<br />

1, 2, 4<br />

patients with AIDS.<br />

III.<br />

Indications of Misc. Antivirals-Foscarnet<br />

Foscarnet sodium has three FDA-approved indications: CMV retinitis in patients with AIDS,<br />

combination therapy with ganciclovir for patients who have relapsed after monotherapy with<br />

either drug, and HSV infections. Although non-FDA approved indications, foscarnet has been<br />

used for colitis, CMV prophylaxis, esophagitis, gastroenteritis, herpes zoster, and varicella-zoster<br />

virus.<br />

Table 2. FDA-Approved Indications 4, 5<br />

Drug<br />

Approved Indications<br />

Foscarnet 1. CMV Retinitis<br />

2. Relapsed CMV Retinitis<br />

3. Mucocutaneous Acyclovir-Resistant HSV Infections<br />

IV.<br />

Pharmacokinetic Parameters of the Misc. Antivirals-Foscarnet<br />

Due to foscarnet’s poor oral bioavailability, the drug is administered by intravenous infusion.<br />

Foscarnet accumulates in bone and is distributed into cartilage. The foscarnet terminal half-life is<br />

determined by urinary excretion, was 87.5+ 41.8 hours, possibly because of release of foscarnet<br />

from bone. Uptake of foscarnet into the bone matrix is likely due to its structural similarity to<br />

phosphate. Penetration into the cerebrospinal fluid has also been observed at concentrations that<br />

are 10% to 50% of plasma levels. CSF penetration is increased with meningeal inflammation. It<br />

is unknown whether the agent crosses the placenta or is distributed into breast milk. Foscarnet is<br />

not metabolized in the liver. Foscarnet exhibits triphasic kinetics. In patients with normal renal<br />

function, the distribution and elimination half-lives are 0.4-1.4 and 3.3-6.8 hours, respectively.<br />

The plasma half-life can be prolonged to up to eight hours in patients with creatinine clearances of<br />

44-90ml/min (see Dosage). The terminal half-life is estimated at 18-88 hours. Between 79-92%<br />

of a dose is eliminated unchanged in the urine through glomerular filtration and tubular secretion.<br />

Tubular reabsorption can occur. 1, 5<br />

255


Foscarnet<br />

Foscarnet selectively<br />

inhibits the viralspecific<br />

DNA<br />

polymerases and<br />

reverse transcriptases at<br />

the pyrophosphatebinding<br />

site. This<br />

occurs through<br />

prevention of<br />

pyrophosphate cleavage<br />

from deoxynucleoside<br />

triphosphate and<br />

elongation of the viral<br />

DNA chain. Foscarnet<br />

also inhibits HIV<br />

reverse transcriptase<br />

and hepatitis B DNA<br />

polymerase.<br />

12-22% 14-17% Biotransformation<br />

does not occur<br />

Active<br />

Metabolit<br />

es<br />

N o<br />

Elimination<br />

urinary<br />

excretion<br />

< 28%<br />

unchanged<br />

drug<br />

Table 3. Pharmacokinetic Parameters of the Misc. Antivirals-Foscarnet 1, 2<br />

Drug Mechanism of Action Bioavailability<br />

Protein<br />

Binding<br />

Metabolism<br />

Half-<br />

Life<br />

3<br />

hours<br />

V. Drug Interactions with the Misc. Antivirals-Foscarnet<br />

Table 4 lists the most documented (Level 1 and 4) drug-drug interactions for foscarnet. One level<br />

1 interaction is documented with foscarnet.<br />

Table 4. Drug Interactions of the Misc. Antivirals-Foscarnet 6<br />

Drug Significance Interaction Mechanism<br />

Foscarnet<br />

Level 1<br />

delayed, major, suspected<br />

Foscarnet and cyclosporin The risk of renal failure may be increased due to possible additive<br />

or synergistic nephrotoxicity.<br />

Level 4<br />

moderate, possible<br />

Foscarnet and quinolones The risk of seizure may be increased due to t he possible<br />

additive or synergistic epileptogenic activity.<br />

Additional less severe drug-drug interactions have been documented with foscarnet and include<br />

the following drugs 1, 6 :<br />

• Nephrotoxic drugs (e.g., aminoglycosides, amphotericin B, IV pentamidine) - Because of<br />

foscarnet’s tendency to cause renal impairment, avoid the use of foscarnet in combination<br />

with potentially nephrotoxic drugs, unless the potential benefits outweigh the risks to the<br />

patient.<br />

• Pentamidine IV - Penicillins (Level 4) - Concomitant treatment with foscarnet and IV<br />

pentamidine may have caused hypocalcemia, hypomagnesemia, and nephrotoxicity.<br />

Toxicity associated with concomitant use of aerosolized pentamidine has not been<br />

reported.<br />

• Zidovudine – Concurrent use of foscarnet and zidovudine may be associated with a<br />

potentiation of anemia.<br />

256


1, 4, 5<br />

VI. Adverse Drug Events of the Misc. Antivirals-Foscarnet<br />

Foscarnet use is associated with a black box warning with the following important prescribing<br />

information.<br />

1, 4, 5<br />

Black Box Warning<br />

Renal Impairment is the major toxicity of foscarnet. Continual assessment of a patient’s risk,<br />

frequent monitoring of serum creatinine with dose adjustment for changes in renal function and<br />

adequate hydration with administration are imperative (see administration and dosage).<br />

Seizures, related to alternations in plasma minerals and electrolytes, have been associated with<br />

foscarnet treatment. Therefore, patients must be carefully monitored for such changes and their<br />

potential sequelae. Mineral and electrolyte supplementation may be required.<br />

Foscarnet is indicated for the use only in immunocompromised patients with CMV retinitis and<br />

mucocutaneous acyclovir-resistant HSV infections (see indications).<br />

Note<br />

Due to the complex nature of the disease for which foscarnet is intended, differentiating between<br />

drug-induced adverse reactions from manifestations of the underlying disease process, is difficult.<br />

GU<br />

Renal function alterations, including increased serum creatinine, and abnormal renal function (see<br />

Warnings) occur in > 5% of patients. Patients differ in degree of foscarnet-induced renal<br />

dysfunction (i.e., change from baseline creatinine clearance). Patients with pre-existing renal<br />

disease should be closely monitored for renal toxicity. Following specific dosage guidelines for<br />

renal impairment is imperative (see Dosage). Baseline renal impairment has been associated with<br />

seizures occurring during treatment with foscarnet. Toxic nephropathy and uremia are rare (


other than electrolyte imbalance that have precipitated seizures in patients receiving foscarnet<br />

include a low baseline absolute neutrophil count and baseline renal impairment.<br />

Respiratory<br />

Respiratory complications, such as cough and dyspnea, have been experienced by at least 5% of<br />

patients receiving foscarnet. More serious but less frequently reported effects include pneumonia,<br />

pulmonary infiltration, pneumothorax, and hemoptysis (1-5%). Pulmonary hemorrhage is rare<br />

(


1, 2, 4<br />

VII. Dosing and Administration of the Misc. Antivirals-Foscarnet<br />

Table 6. Dosing and Administration for Foscarnet<br />

Drug Availability Dose /Frequency/Duration<br />

Foscarnet<br />

Sodium<br />

Foscarnet<br />

solution for<br />

injection<br />

24mg/ml<br />

Caution:<br />

Do not administer by rapid or bolus IV injection. Toxicity may be increased as a result of<br />

excessive plasma levels. Take care to avoid unintentional overdose; carefully control the<br />

rate of infusion by using an infusion pump. In spite of the use of an infusion pump,<br />

overdoses have occurred.<br />

Hydration:<br />

Hydration may reduce the risk of nephrotoxicity. It is recommended that 750 to 1000ml of<br />

normal saline or 5% dextrose solution should be given prior to the first infusion of foscarnet<br />

to establish diuresis. With subsequent infusions, 750 to 1000ml of hydration fluid should be<br />

given with 90 to 120mg/kg of foscarnet and 500ml with 40 to 60mg/kg of foscarnet.<br />

Hydration fluid may need to be decreased if clinically warranted. After the first dose, the<br />

hydration fluid should be administered concurrently with each infusion of foscarnet.<br />

Administration:<br />

Administer by controlled IV infusion, either by using a central venous line or a peripheral<br />

vein. The standard 24mg/ml solution may be used without dilution when using a central<br />

venous catheter for infusion. When a peripheral vein catheter is used, dilute the 24mg/ml<br />

solution to 12mg/ml with 5% dextrose in water or with a normal saline solution prior to<br />

administration to avoid local irritation of peripheral veins. Because the dose is calculated on<br />

the basis of body weight, it may be desirable to remove and discard any unneeded quantity<br />

from the bottle before starting with the infusion to avoid overdosage.<br />

Induction treatment:<br />

CMV retinitis:<br />

The recommended initial dose for patients with normal renal function is either 90mg/kg (1.5-<br />

to 2-hour infusion) every 12 hours or 60mg/kg over a minimum of one hour every eight<br />

hours for 2 to 3 weeks depending on clinical response.<br />

HSV infections:<br />

The recommended initial dose for acyclovir-resistant HSV patients with normal renal<br />

function is 40mg/kg (minimum 1 hour infusion) either every 8 or 12 hours for 2 to 3 weeks<br />

or until healed.<br />

An infusion pump must be used to control the rate of infusion. Adequate hydration is<br />

recommended to establish diuresis, both prior to and during treatment to minimize renal<br />

toxicity, provided there are no clinical contraindications.<br />

Maintenance treatment:<br />

90 to 120mg/kg/day (individualized for renal function) given as an IV infusion over two<br />

hours. Because the superiority of the 120mg/kg/day has not been established in controlled<br />

trials and given the likely relationship of higher plasma foscarnet levels to toxicity, it is<br />

recommended that most patients be started on maintenance treatment with a dose of<br />

90mg/kg/day. Escalation to 120mg/kg/day may be considered should early reinduction be<br />

required because of retinitis progression. Some patients who show excellent tolerance to<br />

foscarnet may benefit from initiation of maintenance treatment at 120mg/kg/day earlier in<br />

their treatment. An infusion pump must be used to control the rate of infusion with all doses.<br />

Again, hydration to establish diuresis both prior to and during treatment is recommended to<br />

minimize renal toxicity.<br />

Patients who experience progression of retinitis while receiving maintenance therapy may be<br />

retreated with the induction and maintenance regimens given above.<br />

259


Special Dosing Considerations<br />

1, 4, 5<br />

Table 7a. Special Dosing Considerations for the Misc. Antivirals-Foscarnet<br />

Drug Renal Dosing Pediatric Use Pregnancy<br />

Category<br />

Foscarnet<br />

Renal function impairment:<br />

Use with caution in patients with<br />

abnormal renal function because reduced<br />

plasma clearance of foscarnet will result<br />

in elevated plasma levels. In addition,<br />

foscarnet has the potential to further<br />

impair renal function. Safety and efficacy<br />

data for patients with baseline serum<br />

creatinine levels> 2.8mg/dL or measured<br />

24-hour creatinine clearances < 50ml/min<br />

are limited. Carefully monitor renal<br />

function at baseline and during induction<br />

and maintenance therapy with appropriate<br />

dose adjustments. If CCl falls below the<br />

limits of the dosing nomograms<br />

(0.4ml/min/kg) during therapy,<br />

discontinue foscarnet and monitor the<br />

patient daily until resolution of renal<br />

impairment is ensured.<br />

The safety and<br />

effectiveness in<br />

pediatric<br />

patients has not<br />

been<br />

established.<br />

C<br />

Can Drug Be<br />

Crushed/Stability<br />

At a concentration of<br />

12mg/ml in normal<br />

saline solution,<br />

foscarnet is stable for<br />

30days a 5 o C<br />

Dose adjustment:<br />

Individualize foscarnet dosing according<br />

to the patient's renal function status.<br />

Refer to table 7b for recommended doses<br />

and adjust the dose as indicated.<br />

1, 4, 5<br />

Table 7b. Foscarnet Renal Impairment Dosing-Induction<br />

Foscarnet Dosing Guide Based on CCl for Induction<br />

HSV: Equivalent to<br />

CMV: Equivalent to<br />

CCl<br />

(ml/min/kg)<br />

40mg/kg q 12<br />

hr<br />

40mg/kg q 8<br />

hr<br />

60mg/kg q 8<br />

hr<br />

90mg/kg q 12<br />

hr<br />

> 1.4 40 q 12 hr 40 q 8 hr 60 q 8 hr 90 q 12 hr<br />

> 1 to 1.4 30 q 12 hr 30 q 8 hr 45 q 8 hr 70 q 12 hr<br />

> 0.8 to 1 20 q 12 hr 35 q 12 hr 50 q 12 hr 50 q 12 hr<br />

> 0.6 to 0.8 35 q 24 hr 25 q 12 hr 40 q 12 hr 80 q 24 hr<br />

> 0.5 to 0.6 25 q 24 hr 40 q 24 hr 60 q 24 hr 60 q 24 hr<br />

> 0.4 to 0.5 20 q 24 hr 35 q 24 hr 50 q 24 hr 50 q 24 hr<br />

< 0.4 Not<br />

Not<br />

Not<br />

Not<br />

recommended recommended recommended recommended<br />

260


Table 7c. Foscarnet Renal Impairment Dosing-Maintenance<br />

Foscarnet Dosing Guide Based on Ccr for<br />

Maintenance<br />

CMV: Equivalent to<br />

90mg/kg/day 120mg/kg/day<br />

CCl<br />

(ml/min/kg)<br />

> 1.4 90 q 24 hr 120 q 24 hr<br />

> 1 to 1.4 70 q 24 hr 90 q 24 hr<br />

> 0.8 to 1 50 q 24 hr 65 q 24 hr<br />

> 0.6 to 0.8 80 q 48 hr 105 q 48 hr<br />

> 0.5 to 0.6 60 q 48 hr 80 q 48 hr<br />

> 0.4 to 0.5 50 q 48 hr 65 q 48 hr<br />

< 0.4 Not<br />

Not<br />

recommended recommended<br />

VIII.<br />

Comparative Effectiveness of the Misc. Antivirals-Foscarnet<br />

Table 8 describes clinical studies that have recently looked at the efficacy and place-in-therapy of<br />

foscarnet.<br />

Table 8. Additional Outcomes Evidence for Foscarnet<br />

Study Sample Treatment /<br />

Duration<br />

A randomized,<br />

controlled trial<br />

comparing<br />

ganciclovir to<br />

ganciclovir plus<br />

foscarnet ( each<br />

at half dose) for<br />

preemptive<br />

therapy of<br />

cytomegalovirus<br />

infection in<br />

transplant<br />

recipients. 7 n=48 Full-dose ganciclovir<br />

(5mg/kg IV twice daily)<br />

versus half-dose<br />

ganciclovir(5mg/kg IV<br />

once daily) and halfdose<br />

foscarnet(90mg/kg<br />

IV once daily/<br />

14 days<br />

261<br />

Results<br />

The goal of this randomized controlled trail was to determine the effectiveness<br />

and safety of full-dose ganciclovir (5mg/kg IV twice daily) versus half-dose<br />

ganciclovir (5mg/kg IV once daily) and half-dose foscarnet (90mg/kg IV once<br />

daily):<br />

• In the ganciclovir arm, 17 of the 24 patients reached the endpoint of being<br />

CMV negative by PCR within 14 days of therapy.<br />

• The half-dose ganciclovir and half-dose foscarnet arm, 12 of the 24 patients<br />

reached the endpoint of being CMV negative by PCR within 14 days of<br />

therapy.<br />

• Toxicity was greater in the combination therapy arm.<br />

• Summary: The study did not support the synergistic effect of ganciclovir<br />

plus foscarnet in vivo.<br />

n=48 Foscarnet versus<br />

Treatment of<br />

In examining the clinical effectiveness of the two different antivirals for<br />

randomized<br />

• Further evidence of CMV disease was seen in 35% of patients during<br />

comparison. 8 followup; time to progression was not significantly different between<br />

AIDS-associated<br />

Gastrointestinal<br />

ganciclovir infection with CMV, in terms visual analog score of symptoms, endoscopic<br />

appearances, histologic inflammation, and number of CMV inclusions:<br />

cytomegalovirus<br />

• In each treatment group, 73% has good clinical response.<br />

infection with<br />

foscarnet and<br />

ganciclovir: a<br />

• Foscarnet treated group showed 83% response and Ganciclovir treated<br />

group showed 85% response by endoscopy; Inclusion bodies disappeared<br />

from follow-up biopsies in 73% of these.<br />

recipients.<br />

• Summary: Both ganciclovir and foscarnet are effective first line agents for<br />

gastrointestinal CMV infection. Maintenance therapy does not prevent<br />

progression of disease.<br />

Combination n=279 Three therapeutic In examining the mortality, retinitis progression, visual acuity, visual fields, and


foscarnet and<br />

ganciclovir<br />

therapy vs<br />

monotherapy for<br />

the treatment of<br />

relapsed<br />

cytomegalovirus<br />

retinitis in<br />

patients with<br />

AIDS. The<br />

Cytomegalovirus<br />

Retreatment<br />

Trial. The Studies<br />

of Ocular<br />

complication of<br />

AIDS Research<br />

Group in<br />

Collaboration<br />

with the AIDS<br />

Clinical Trials<br />

Group. 9<br />

regimens:<br />

* foscarnet group:<br />

induction with foscarnet<br />

at 90mg/kg IV q 12 hr<br />

for 2 weeks, followed<br />

by a maintenance dose<br />

of 120mg/kg per day<br />

* ganciclovir group:<br />

induction with<br />

ganciclovir at 5mg/kg<br />

IV q 12 hr for two<br />

weeks followed by<br />

maintenance at<br />

10mg/kg per day<br />

* continuation of<br />

previous maintenance<br />

therapy plus induction<br />

with the other drug<br />

(ganciclovir or<br />

foscarnet) for two<br />

weeks followed by<br />

maintenance therapy<br />

with both drugs<br />

(ganciclovir 5mg/kg per<br />

day and foscarnet at<br />

90mg/kg per day.<br />

morbidity:<br />

• The mortality rate was similar among the three groups: foscarnet group 8.4<br />

months, ganciclovir group 9.0 months, and combination therapy group 8.6<br />

months (P=.89).<br />

• Retinitis progression (determined by the Fundus Photograph Reading<br />

Center revealed that the combination therapy was the most effective<br />

regimen for controlling the retinitis. The median time to regression:<br />

foscarnet group 1.3 months, ganciclovir group 2.0 months, and combination<br />

therapy group 4.3 months (P=.001).<br />

• No difference was seen on the visual acuity outcomes.<br />

• The rates of visual field loss were: foscarnet group 28 degrees per month,<br />

ganciclovir group 18 degrees per month, and combination therapy group 16<br />

degrees per month (P=.009).<br />

• Rates of increased retinal area involved by CMV were as follows: foscarnet<br />

group 2.47% per month, ganciclovir group 1.4% per month, and<br />

combination therapy group 1.19% per month (P=.009).<br />

• Side effects were similar, but the combination group had the greatest<br />

negative impact of treatment on the quality of life measure.<br />

• Summary: For patients with AIDS and CMV retinitis whose retinitis has<br />

relapsed and can tolerate both drugs, combination therapy appears to be the<br />

most effective therapy to control CMV retinitis.<br />

Foscarnet vs<br />

ganciclovir for<br />

Cytomegalovirus<br />

(CMV)<br />

antigenemia after<br />

allogenic<br />

hemopoietic stem<br />

cell<br />

transplantation<br />

(HSCT): a<br />

randomized<br />

study 10 n=39 15 days The goal of this trial was designed to compare foscarnet with ganciclovir as preemptive<br />

therapy for CMV infection in patients undergoing hemopoitic stem cell<br />

transplantation. Patients were randomized to receive foscarnet 90mg/kg q 12h vs<br />

ganciclovir 5mg/kg q12h for 15 days at the time of development of CMV Agemia.<br />

The study was designed to determine outcome of CMVAg-emia,<br />

progression to CMV disease and side effects of treatment. Increments of serum<br />

creatinine in the foscarnet group and cytopenia in the ganciclovir group were<br />

controlled by reducing the administered dose.<br />

• Clearance of CMVAg-emia was faster in the foscarnet group.<br />

• Failures of treatment occurred in 3/20 patients in foscarnet group vs 8/19<br />

patients in ganciclovir group.<br />

• Summary: Foscarnet and ganciclovir are effective for pre-emptive therapy<br />

of CMVAg-emia. Side effects can be managed with appropriate dose<br />

reduction.<br />

Foscarnet<br />

n=26 - Mucocutaneous herpes simplex virus in 26 patients was studied after patients<br />

mucocutaneous<br />

• Cessation of viral shedding was noted in all of the 11 patients that were<br />

herpes simplex<br />

recultured.<br />

virus infection in<br />

• The 14 patients that received vidarabine for acyclovir resistant HSV before<br />

26 AIDS patients:<br />

the foscarnet therapy did not see their HSV resolve.<br />

preliminary data 11<br />

treatment of<br />

received foscarnet.<br />

acyclovirresistant<br />

• Clinical response was noted in 81% of patients<br />

• Complete reepithelialization of HSV lesions occurred in 73% of<br />

patients.<br />

In vitro<br />

susceptibility<br />

study of herpes<br />

simplex virus<br />

acyclovir and<br />

foscarnet. Are<br />

routine<br />

susceptibility<br />

studies<br />

necessary 12 n=68 - Objective of study was to determine the prevalence of resistance of herpes<br />

simplex virus to acyclovir and foscarnet.<br />

• An in vitro susceptibility study of HSV strains isolated from HIV –infected<br />

and non-HIV infected patients was conducted by means of qualitative<br />

screening.<br />

• Results included investigating 84 HSV strains, 49 HIV-infected patients<br />

and 19 control patients. No strains in the control group were resistant to<br />

acyclovir. In HIV-infected patients one acyclovir resistant strain was<br />

to<br />

detected and one moderately resistant to acyclovir strain was detected. The<br />

moderately resistant strain had good response to acyclovir treatment. No<br />

resistance to foscarnet was detected.<br />

• Summary: Percentages to HSV strains resistant to acyclovir is low and<br />

resistance to foscarnet was not detected. Study suggests that routine in-vitro<br />

susceptibility testing of antiviral drugs used against HSV does not seem to<br />

262


e necessary.<br />

Additional Evidence<br />

Dose Simplification: Since foscarnet is only available in a single formulation, there isn’t another<br />

agent or dosage formulation for which it can be compared to for dose simplification. Therefore,<br />

dose simplification does not apply to this particular class.<br />

Stable Therapy: Limited clinical data is available regarding changing from one antiviral agent<br />

for CMV to another agent, and the impact on the outcome of the disease. One study by Flores-<br />

Aguilar M et al. looked at the treatment of clinically resistant CMV retinitis. 13 In a study group of<br />

100 patients, 11 developed clinically resistant retinitis, despite at least eight weeks of induction<br />

doses of either foscarnet or ganciclovir. Therapeutic interventions for resistant infections included<br />

continuing high dose of the same antiviral drug, changing to an alternative antiviral agent, or<br />

combining antiviral therapy with foscarnet and ganciclovir. The use of combination therapy with<br />

foscarnet and ganciclovir was effective in halting progession of retinitis in three (75%) of four<br />

patients receiving combination therapy.<br />

Impact on Physician Visits: Not applicable.<br />

IX.<br />

Conclusions<br />

Use of foscarnet should be reserved for approved infections or infections resistant to other<br />

therapies. Use is likely limited to hospitalized patients, where intravenous therapy can be given<br />

and monitored appropriately. Because of the black box warning associated with foscarnet use, and<br />

the fact that the drug has limited indications, use of foscarnet is not applicable to the general<br />

population. Foscarnet should be available for such special needs/circumstances that require<br />

medical justification through the prior authorization program.<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the<br />

generics in the class and offer no significant clinical advantage over other alternatives in general<br />

use.<br />

X. Recommendations<br />

No brand of foscarnet is recommended for preferred status.<br />

263


References<br />

1. Clinical Pharmacology, 2004. Available on-line at: www.cp.gsm.com. Accessed September<br />

2004.<br />

2. McEvoy GK, Ed. American Hospital Formulary Service, AHFS Drug Information. American<br />

Society of Health-System Pharmacists. Bethesda. 2004.<br />

3. World Health Organization. WHO essential Medicines Library. WHO practical guidelines. 2 nd<br />

edition. Available at: www.who.int/emc. Accessed September 20, 2004.<br />

4. Astra-Zeneca. Foscavir package insert<br />

5. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

6. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

7. Mattes FM, Hainworth EG, Geretti AM, et al. A randomized, controlled trail comparing<br />

ganciclovir to ganciclovir plus foscarnet (each at half dose) for preemptive therapy of<br />

cytomegalovirus infection in transplant recipients. J Infect Dis. 2004 Apr;189(8): 1355-61.<br />

8. Chelsa and Westminister Hospital. Treatment of AIDS-associated gastrointestinal<br />

cytomegalovirus infection with foscarnet and ganciclovir: a randomized comparison. J Infect Dis.<br />

1995 Sept;172(3):622-8.<br />

9. No authors listed. Combination foscarnet and ganciclovir therapy vs monotherapy for the<br />

treatment of relapsed cytomegalovirus retinitis in patients with AIDS. The Cytomegalovirus<br />

Retreatment Trial. The Studies of Ocular complication of AIDS Research Group in Collaboration<br />

with the AIDS Clinical Trials Group. Arch ophthalmol. 1996 Jan;114(1):23-33.<br />

10. Moretti S, Zikos P, Van Lint MT et al. Foscarnet vs ganciclovir for Cytomegalovirus (CMV)<br />

antigenemia after allogenic hemopoietic stem cell transplantation (HSCT): a randomized study.<br />

Bone Marrow Transplant. 1998 Jul;22(2):175-80.<br />

11. Safrin S, Assaykeen T, Follansbee S, et al. Foscarnet treatment of acyclovir-resistant<br />

mucocutaneous herpes simplex virus infection in 26 AIDS patients: preliminary data. J Infect Dis.<br />

1990 Jun;161(6):1078-84.<br />

12. Losada I, Canizares A, Hellin T, et al. In vitro susceptibility study of herpes simplex virus to<br />

acyclovir and foscarnet. Are routine susceptibility studies necessary. Enferm Infecc Microbiol<br />

Clin.2002 Jan;20(1): 25-7.<br />

13. Flores-Aguilar M, Kuppermann BD, Quiceno JI, et al. Pathophysiology and treatment of<br />

clinically resistant cytomegalovirus retinitis. Ophthalmology 1993 Jul;100(7):1022-31.<br />

264


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Amebicides<br />

AHFS 083004<br />

October 27, 2004<br />

I. Overview<br />

Amebiasis is a parasitic infection caused by the protozoon Entamoeba histolytica (E. histolytica).<br />

In the United States and other developed countries, infection occurs primarily in immigrants from<br />

underdeveloped regions and travelers returning from underdeveloped regions. Institutionalized<br />

individuals (particularly the mentally retarded), homosexual males, and immunosuppressed<br />

individuals are also at high risk for infection. 1 Other Entamoeba strains that are morphologically<br />

identical to E. histolytica, but are nonpathogenic are Entamoeba dispar, Entamoeba coli, and<br />

Entamoeba hartmanni.<br />

Globally, amebiasis affects approximately 50 million people annually, with the highest prevalence<br />

being in the Indian subcontinent, southern and western Africa, the Far East, South America, and<br />

Central America. Globally, amebiasis causes nearly 100,000 deaths annually and is the third<br />

leading parasitic cause of death. 2 In the United States, prevalence of Entamoeba infection is<br />

approximately 4%, although only 10% of persons who are infected have symptomatic disease.<br />

Asymptomatic cyst carriers can be treated with the same drug regimen as those who have<br />

symptomatic intestinal amebiasis.<br />

The life cycle of E. histolytica involves two forms - cysts and trophozoites. Outside of the human<br />

body, E. histolytica can only survive in cyst form, and is therefore the form in which the organism<br />

is transmitted through contaminated feces, water, or food. Once inside the small intestine, the<br />

trophozoite is released from the cyst and becomes the motile (pseudopod) form. It may then<br />

invade the mucosal cells of the colon and produce ulcerative lesions and in rare cases, invade sites<br />

outside of the colon. However, only 10% of infected persons present with symptoms. In 90% of<br />

cases, the trophozoite will re-encyst due to unfavorable conditions in the colon and the infection<br />

will become asymptomatic. Re-encysted trophozoites may either stay in the bowel, or be released<br />

in the stool, where they can be transmitted to other humans.<br />

E. histolytica infections can remain inside the intestine or they can spread to include abscesses of<br />

the perineum, genitalia, pericardium, peritoneum, liver, lungs, and the brain in rare cases. Clinical<br />

manifestations of intestinal infection range from vague complaints of abdominal discomfort and<br />

malaise to severe abdominal cramps, flatulence, and bloody diarrhea with mucus. 1 Fever occurs<br />

only in 10-30% of patients. Additional, but uncommon manifestations of amebic colitis include<br />

fulminant colitis, ameboma, rectal fistulas, or vaginal fistulas. E. histolytica infections can spread<br />

to the liver via portal blood and cause liver abscess. Right upper quadrant pain, hepatomegaly,<br />

and liver tenderness with referred pain to the right or left shoulder usually suggests amebic liver<br />

abscess. 1 Liver abscesses that are located in the right lobe can spread to the lungs and pleura. 1<br />

Amebic pericarditis and amebic peritonitis are associated with left liver lobe rupture. The onset of<br />

cerebral amebiasis is abrupt and usually leads to death within 12-72 hours.<br />

Diagnosis of amebiasis may include microscopy, stool culture, antigen detection, serology, and<br />

molecular probes. 2 Microscopy and stool culture remain the most common methods of diagnosis<br />

in endemic areas, however it may be difficult to make a specific diagnosis since E. histolytica<br />

cannot be distinguished from E. dispar. Serum antibody detection is most useful in distinguishing<br />

between E. histolytica and other organisms as well as diagnosing extraintestinal disease where<br />

organisms may not be present in stool samples and is most useful in non-endemic areas.<br />

Colonoscopy or sigmoidoscopy may be used to diagnose amebic colitis in order to observe amebic<br />

ulcers and allow for biopsy or aspiration. Imaging studies such as chest radiography, ultrasound,<br />

CT scans, or MRI are useful in diagnosing extraintestinal disease. Another challenge of<br />

diagnosing amebiasis is to distinguish it from other gastrointestinal diseases, such as bacterial<br />

dysentery, inflammatory bowel disease, or ischemic colitis.<br />

265


This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this<br />

review.<br />

Table 1. Amebicides in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name (s)<br />

Paromomycin Sulfate Oral Capsule *Humatin<br />

**Iodoquinol Oral Tablet, Powder Yodoxin, Diquinol<br />

Tinidazole Oral Tablet Tindamax 1<br />

*Generic Available.<br />

**At this time, there are no active iodoquinol products in the Alabama Medicaid drug file database.<br />

1 Tindamax (tinidazole), a new drug, was approved in the summer of 2004. Per Alabama Medicaid P & T policy, tinidazole<br />

is eligible for review after it has been commercially available for at least 6 months. Tinidazole will be reviewed at a future<br />

time.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Paromomycin is indicated for the treatment of acute and chronic intestinal amebiasis and is also<br />

indicated as adjunctive therapy in the management of hepatic coma. 4 Paromomycin has a<br />

spectrum of activity is similar to that of neomycin, but the drug is considered a luminal or contact<br />

amebicide. Paromomycin is not absorbed into systemic circulation, so its use is limited only to<br />

infection within the intestine (luminal infection). In the case of extraintestinal infection, a course<br />

of paromomycin will follow a course of another anti-infective agent, such as metronidazole.<br />

Iodoquinol is indicated for the treatment of intestinal amebiasis. Like paromomycin, iodoquinol is<br />

not absorbed into systemic circulation, so its use is limited only to infection within the intestine<br />

(luminal infection). In the case of extraintestinal infection, a course of iodoquinol will follow a<br />

course of another anti-infective agent, such as metronidazole. The exact mechanism of action of<br />

iodoquinol is unknown, but the drug is active against both the trophozoite and cyst forms of E.<br />

histolytica. 4<br />

The Centers for Disease Control and Prevention have instituted recommendations for preventing<br />

opportunistic infections among immunosuppressed patients such as those infected with human<br />

immunodeficiency virus (HIV) and hematopoietic stem cell transplant recipients (HSCT).<br />

Specifically, the 2001 USPHS/IDSA guidelines for the prevention of opportunistic infections in<br />

persons infected with HIV recommend that patients avoid sexual practices that might result in oral<br />

exposure to feces to reduce the risk for intestinal infections. 6 In addition, patients should take<br />

precautions when traveling to reduce the risk of food-borne and waterborne infections.<br />

Recommendations of the CDC concerning HSCT recipients are very specific in regards to travel<br />

safety, food safety practices, and safe sexual practices in the prevention of all types of<br />

opportunistic infections.<br />

Another luminal agent, diloxanide furoate, is frequently mentioned in the literature available on<br />

the treatment of amebiasis, but is not readily available in the United States, and is therefore not<br />

mentioned in this review.<br />

No additional guidelines were found on the use of amebicides.<br />

266


III.<br />

Comparative Indications of the Amebicide Antibiotics<br />

Table 2 lists indications for the two drugs in the amebicide class. Paromomycin has two FDA-approved indications: acute and chronic amebiasis and as<br />

adjunctive therapy in the treatment of hepatic coma. Iodoquinol has one FDA-approved use for the treatment of acute and chronic intestinal amebiasis.<br />

Table 2. Indications for the Amebicides 4, 8<br />

Drug Entamoeba histolytica Dientamoeba fragilis<br />

Diphyllobothrium latum<br />

Taenia saginata<br />

Taenia solium<br />

Dipylidium caninum<br />

Hymenolepis nana<br />

Paromomycin<br />

Iodoquinol<br />

✔<br />

Acute and chronic<br />

intestinal amebiasis. Not<br />

effective alone for the<br />

treatment of extraintestinal<br />

amebiasis (i.e. liver<br />

abscess), where it must be<br />

combined with an<br />

absorbable anti-infective<br />

such as metronidazole.<br />

✔<br />

Acute and chronic<br />

intestinal amebiasis. Not<br />

effective alone for the<br />

treatment of extraintestinal<br />

amebiasis (i.e. liver<br />

abscess), where it must be<br />

combined with an<br />

absorbable anti-infective<br />

such as metronidazole.<br />

✔ *<br />

Unlabeled use as an<br />

alternative agent in the<br />

treatment of certain<br />

tapeworm infections.<br />

Giardia lamblia<br />

✔ * *<br />

Unlabeled use as an<br />

alternative agent in the<br />

treatment of giardiasis.<br />

L. donovani<br />

L. braziliensis<br />

✔ * * *<br />

Unlabeled use as an<br />

alternative agent in the<br />

treatment of visceral<br />

and cutaneous<br />

leishmaniasis.<br />

Cryptosporidium muris<br />

✔ * * * *<br />

Unlabeled use as an<br />

alternative agent in the<br />

treatment of<br />

cryptosporidiosis in<br />

immunocompromised<br />

patients.<br />

Hepatic coma<br />

- - - - -<br />

✔<br />

Indicated as<br />

adjunctive<br />

therapy in<br />

hepatic coma.<br />

*<br />

Uncontrolled trials suggest that paromomycin may be effective in the treatment of tapeworm infestations. 5<br />

**<br />

Paromomycin can be considered an alternative treatment of giardiasis, particularly in pregnant patients. 5<br />

***<br />

There is evidence that paromomycin may be effective in the treatment of certain forms of leishmaniasis. 5<br />

****<br />

There are favorable but inconsistent reports that paromomycin is useful in the treatment of cryptosporidiosis. 5


IV.<br />

Pharmacokinetic Parameters<br />

Table 4 lists the pharmacokinetic parameters and mechanisms of action of the amebicide<br />

antibiotics.<br />

Table 3. Pharmacokinetic Parameters of the Amebicide Agents 4, 8<br />

Drug Mechanism of Action Bioavailability Protein<br />

Binding<br />

Paromomycin<br />

Iodoquinol<br />

An amebicidal and<br />

antibacterial aminoglycoside.<br />

Antibacterial activity closely<br />

parallels neomycin.<br />

Aminoglycosides acts by<br />

irreversibly binding to the 30S<br />

subunit of bacterial ribosomes,<br />

blocking the recognition step<br />

in protein synthesis and<br />

causing misreading of the<br />

genetic code. The ribosomes<br />

separate from messenger<br />

RNA, which causes death of<br />

the cell.<br />

Amebicidal against<br />

trophozoite and cyst forms of<br />

E. histolytica. Exact<br />

mechanism of action is<br />

unknown.<br />

Poorly absorbed.<br />

Almost 100% of the<br />

drug is excreted in<br />

the stool.<br />

Poorly absorbed<br />

(~8%). Eliminated in<br />

the stool. Less than<br />

10% recovered in the<br />

urine mostly as<br />

glucuronides.<br />

Metabolism Active<br />

Metabolites<br />

Elimination<br />

- - - Biliary;


VI.<br />

Adverse Drug Events of the Amebicidal Agents<br />

The most common adverse reactions to paromomycin are nausea, abdominal cramps, and diarrhea<br />

at does greater than 3gm daily. 4 Inadvertent absorption through ulcerative bowel lesions may<br />

result in eighth cranial nerve damage (ototoxicity) and renal damage. 4 Prolonged or repeated use<br />

of paromomycin may result in bacterial or fungal overgrowth leading to secondary infections. 4<br />

Paromomycin is contraindicated in patients with a history of previous sensitivity reactions and in<br />

patients with intestinal obstruction. 8<br />

The most common adverse reactions to iodoquinol include skin eruptions, urticaria, pruritus,<br />

nausea, vomiting, abdominal cramps, diarrhea, and pruritus ani. 4 Fever, chills, headache, vertigo,<br />

and thyroid enlargement have also been reported. 4 Prolonged high dosage therapy has been<br />

associated with optic neuritis, optic atrophy, and peripheral neuropathy and is therefore why the<br />

drug is not indicated for the treatment of chronic diarrhea. 4 Iodoquinol is contraindicated in<br />

patients with previous history of sensitivity to iodine and 8-hydroxyquinolines (i.e. iodoquinol,<br />

iodochlorhydroxyquin) and in patients with hepatic damage. 10<br />

Specific percentages of adverse events were not available for the amebicidal agents.<br />

4, 5, 10<br />

Table 5. Common Adverse Events Reported for the Amebicidal Agents<br />

Adverse Event Paromomycin Iodoquinol<br />

Body as a Whole<br />

Malaise<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Pruritis ani<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Chills<br />

Fever<br />

Headache<br />

Optic neuritis<br />

Optic atrophy<br />

Peripheral neuropathy<br />

Ototoxicity<br />

- -<br />

- -<br />

b<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

-<br />

Skin and Appendages<br />

Skin eruptions<br />

Pruritus<br />

Urticaria<br />

-<br />

-<br />

-<br />

b<br />

b<br />

b<br />

Renal<br />

- -<br />

Renal damage<br />

Other<br />

Thyroid enlargement - b<br />

bAdverse event reported; specific percentages not available<br />

269


VII.<br />

Dosing and Administration for the Amebicidal Agents<br />

Table 6. Dosing for the Amebicidal Agents 4, 5<br />

Drug Availability Dose /Frequency/Duration<br />

Paromomycin<br />

Sulfate<br />

250mg oral capsule Intestinal Amebiasis (symptomatic luminal infection or asymptomatic cystcarriers)<br />

Adults and children: 25 to 35mg/kg/day in 3 divided doses with meals for 5 to<br />

10 days.<br />

Hepatic coma<br />

Adults: 4gm daily in divided doses administered at regular intervals for 5 to 6<br />

days.<br />

Iodoquinol<br />

210mg oral tablet,<br />

650mg oral tablet<br />

Other Notes<br />

• Since paromomycin is not well absorbed, patients with extraintestinal<br />

amebiasis should be treated with another anti-infective first (such as<br />

metronidazole) to eradicate extraintestinal infection, followed by a<br />

course of paromomycin to eradicate luminal infection.<br />

• Asymptomatic cyst carriers can be treated like patients with<br />

symptomatic infection to eradicate cysts which could eventually be<br />

passed on and infect others. Patients infected with non-pathologic<br />

strains of Entamoeba, such as E. dispar, do not need to be treated.<br />

• Unlabeled uses: Dientamoeba fragilis (25 to 30 mg/kg/day in 3 doses<br />

for 7 days). Diphyllobothrium latum, Taenia saginata, Taenia solium,<br />

and Dipylidium caninum (adults: 1gm every 15 minutes for 4 doses;<br />

pediatric: 11mg/kg every 15 minutes for 4 doses). Hymenolepis nana<br />

(45mg/kg/day for 5 to 7 days).<br />

Intestinal Amebiasis (symptomatic luminal infection or asymptomatic cystcarriers)<br />

Adults: 650mg 3 times daily after meals for 20 days.<br />

Children: 40mg/kg daily (maximum 650mg per dose) in 3 divided doses for 20<br />

days. Do not exceed 1.95gm in 24 hours for 20 days.<br />

Other Notes<br />

• Since iodoquinol is not well absorbed, patients with extraintestinal<br />

amebiasis should be treated with another anti-infective first (such as<br />

metronidazole) to eradicate extraintestinal infection, followed by a<br />

course of iodoquinol to eradicate luminal infection<br />

• Asymptomatic cyst carriers can be treated like patients with<br />

symptomatic infection to eradicate cysts which could eventually be<br />

passed on and infect others. Patients infected with non-pathologic<br />

strains of Entamoeba, such as E. dispar, do not need to be treated.<br />

270


Special Dosing Considerations<br />

5, 16<br />

Table 7. Special Dosing Considerations for the Amebicidal Agents<br />

Drug Renal Hepatic Pediatric Use<br />

Paromomycin<br />

Sulfate<br />

Dosing<br />

Dosing<br />

No No Same dosing schedule as<br />

adults.<br />

Iodoquinol No No Although dosing in children is<br />

well established, children may<br />

be more susceptible to the side<br />

effects of optic atrophy, optic<br />

neuritis, and peripheral<br />

neuropathy, especially with<br />

prolonged high-dose therapy.<br />

Pregnancy<br />

Category<br />

C - Limited<br />

information is<br />

available, but the<br />

drug is not well<br />

absorbed, therefore<br />

teratogenic effect is<br />

minimized.<br />

C - Safety for use<br />

in pregnancy has<br />

not been<br />

established.<br />

Can Drug Be Crushed<br />

Information regarding the<br />

opening of paromomycin sulfate<br />

capsules is not available, per the<br />

manufacturer.<br />

Tablets may be crushed and<br />

mixed with food such as<br />

applesauce or chocolate syrup if<br />

unable to swallow.<br />

271


VIII.<br />

Comparative Effectiveness of the Amebicidal Agents<br />

In a search of Medline and Ovid, no recent studies were found comparing the efficacy of<br />

paromomycin versus iodoquinol. Both agents are accepted as the standard treatment of luminal<br />

amebic infections. Most recent studies evaluate the effectiveness of paromomycin in treating<br />

conditions other than intestinal amebiasis, such as leishmaniasis, giardiasis, trichomoniasis,<br />

tapeworm infestation, and cryptosporidiosis in HIV-positive patients, which are currently<br />

unlabeled uses for paromomycin. A few of the larger studies are described in Table 8. Very small<br />

studies and case reports are not listed. In addition, a few microbiological studies were found<br />

suggesting the possibility of E. histolytica resistance to current treatment regimens. A summary of<br />

the most pertinent one can be found in Table 8.<br />

Sullam, N=114 7 day treatment with<br />

al. 12 intestinal amebiasis<br />

PM, et<br />

paromomycin for<br />

White<br />

AC, et<br />

al. 13 n=10 Paromomycin 25-<br />

30mg/kg/day vs.<br />

placebo for 14 days<br />

for cryptosporidiosis<br />

Hewitt<br />

RG, et<br />

al. 14 n=35 Paromomycin 500mg<br />

4 times daily for 42<br />

days vs. placebo for<br />

21 days followed by<br />

paromomycin 500mg<br />

4 times daily for 21<br />

days for<br />

cryptosporidiosis<br />

El-on J, et<br />

al. 15 n=39 Ointment containing<br />

15% paromomycin<br />

and 12% or 5%<br />

methylbenzothonium<br />

chloride<br />

Table 8. Additional Outcomes Evidence for the Amebicides<br />

Study Sample Treatment /<br />

Results<br />

Duration<br />

Samuelso<br />

n, et al. 11 - - Three sets of experiments were performed to better characterize the multi-drug<br />

resistant phenotype of the emetine-resistant amebae. Emetine is the oldest drug used<br />

to treat amebic infection. Mutant amoebae were cross-resistant to certain drugs used<br />

to treat luminal amebiasis (iodoquinol), but not those used to treat systemic infection.<br />

This, combined with isolated reports of failed treatment courses, suggest that drugresistant<br />

amebae may develop over time, which warrants further investigation.<br />

114 homosexual men were evaluated who had mild-to-moderate intestinal amebiasis.<br />

All patients received 25-35 mg/kg daily in three divided doses for 7 days. The results:<br />

• Of the 80 patients with gastrointestinal complaints at the onset of therapy,<br />

55 (80%) of 69 were asymptomatic within 4 to 6 weeks after completion of<br />

treatment and 11 were lost to follow-up.<br />

• Paromomycin produced long-term eradication of intestinal E. histolytica<br />

infection in 92% of all men evaluated. The rate of microbiologic cure<br />

among patients with symptoms at onset of therapy was comparable to that<br />

of asymptomatic individuals.<br />

• Paromomycin was well tolerated, with mild diarrhea during therapy the<br />

only frequent adverse effect (67%).<br />

In a double-blind trial, 10 patients with AIDS and cryptosporidiosis were placed on<br />

either paromomycin 25-30mg/kg/day for 14 days and then switched to placebo, or<br />

vice versa:<br />

• During the paromomycin treatment phase, oocyst excretion and stool<br />

frequency both decreased more in the treatment phase than in the placebo<br />

phase (P


Additional Evidence<br />

Dose Simplification: In most cases, the amebicidal agents in this class are given for a brief<br />

duration (acute use). Long-term high dose therapy is not recommended for either of the two drugs<br />

in this class. Therefore, research into dose simplification within this class is not applicable.<br />

Stable Therapy: Limited clinical data is available on resistance to the amebicidal medications in<br />

humans. Most of the articles available were from a microbiological standpoint suggesting that<br />

resistance is a potential problem that warrants closer examination. Although the clinical evidence<br />

does not suggest problems with resistance patterns in the U.S, resistance to the amebicidal drugs<br />

should be considered in patients who have previously been treated for amebic infections and in<br />

those with apparent treatment failure.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal clinical<br />

literature relevant to use of the amebicides and their impact on physician visits.<br />

IX.<br />

Conclusions<br />

Parasitic infections caused by the organism E. histolytica present in three ways: asymptomatic<br />

intestinal infection, symptomatic intestinal infection, and extraintestinal infection. Asymptomatic<br />

intestinal infection and mild symptomatic intestinal infection are both treated with a course of a<br />

luminal amebicide (paramomycin or iodoquinol). At this time, iodoquinol is not available in the<br />

Alabama Medicaid drug file. Extraintestinal infection (outside of the intestinal lumen) and severe<br />

intestinal infection (amebic dysentery) must be treated with a tissue agent. 2 Metronidazole is the<br />

drug of choice, followed by a course of one of the luminal agents to eradicate organisms still<br />

remaining in the intestine.<br />

No studies were found that compared the efficacy of paromomycin and iodoquinol directly in the<br />

treatment of intestinal amebiasis. Both are accepted as standard, equally efficacious treatment in<br />

patients with asymptomatic intestinal infection and mild symptomatic intestinal infection. There<br />

is more information available about the safety of paromomycin in the use of pregnant women,<br />

although both drugs belong to pregnancy category C. 2, 5<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the<br />

generics in the class and offer no significant clinical advantage over other alternatives in general<br />

use.<br />

X. Recommendations<br />

No brand amebicide is recommended for preferred status.<br />

273


References<br />

1. Anandan JV. Parasitic Diseases. In: <strong>Pharmacotherapy</strong>. A Pathophysiologic Approach. Dipiro<br />

JT, Talbert RL, Hayes, PE, et al. Eds. Elsevier Science Publishing. New York. 1989. Pg. 1194-<br />

1195.<br />

2. Ayeh-Kumi, PF, Petri, WA, et al. Diagnosis and Management of Amebiasis. Infect Med 2002<br />

Oct:19(8):375-382.<br />

3. World Health Organization (WHO) Infectious Diseases. www.who.int accessed September 2004.<br />

4. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

5. Micromedex, Healthcare Series;DrugDex Drug Evaluations 2004.<br />

6. 2001 USPHS/IDSA guidelines for the prevention of opportunistic infections in persons infected<br />

with human immunodeficiency virus. Rockville (MD): U.S. Department of Health and Human<br />

Services, Public Health Service;2001 Nov 28.<br />

7. Centers for Disease Control and Prevention (CDC). Guidelines for preventing opportunistic<br />

infections among hematopoietic stem cell transplant recipients. MMWR Oct 20 2000;49;RR-10.<br />

8. Humatin package insert; Monarch Pharmaceuticals Rev. June 2001<br />

9. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

10. Yodoxin package insert; Glenwood, LLC Rev. January 2003<br />

11. Samuelson, JC, Burke, A, Courval, JM. Susceptibility of an Emetine-Resistant Mutant of<br />

Entamoeba histolytica to Multiple Drugs and to Channel Blockers. Antimicrobial Agents and<br />

Chemotherapy Nov, 1992; 36(11):2392-2397.<br />

12. Sullam PM, Slutkin G, Gottlieb AB, et al. Paromomycin therapy of endemic amebiasis in<br />

homosexual men. Sex Transm Dis. 1986 Jul-Sept;13(3):151-155.<br />

13. White AC, Jr, Chappell, CL, Hayat CS, et al. Paromomycin for cryptosporidiosis in AIDS: a<br />

prospective, double-blind trial. J Infec Dis 1994 Aug;170(2):419-424.<br />

14. Hewitt RG, Yiannoutsos CT, Higgs, ES, et al. Paromomycin: no more effective than placebo for<br />

treatment of cryptosporidiosis in patients with advanced human immunodeficiency virus infection.<br />

Clin Infec Dis.2000 Oct;31(4):1084-1092.<br />

15. El-on J, Halevy S, Grunwald MH, et al. Topical treatment of Old World cutaneous leishmaniasis<br />

caused by leishmania major: a double-blind control study. J Am Acad Dermatol 1992 Aug;27(2 pt<br />

1):227-231.<br />

16. Horga MA. EMedicine – Amebiasis. Updated July 1, 2004. Accessed at<br />

www.emedicine.com/ped/topic80.htm September 2004.<br />

274


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Antimalarial Agents<br />

AHFS 083008<br />

October 27, 2004<br />

I. Overview<br />

Globally, malaria continues to be one of the most important and devastating infectious diseases in<br />

developing regions of the world. More than 40% of the world’s population lives in areas where<br />

malaria transmission occurs, resulting in an estimated 300-500 million cases of malaria each year<br />

attributing to 750,000-2 million deaths. 1<br />

While the transmission of malaria was eradicated in the United States in the 1950s, the threat of<br />

malaria is still a concern due to the number of U.S. travelers who visit areas where malaria is<br />

present. Travelers returning from malaria-endemic countries of the world are the vast majority of<br />

diagnosed cases of malaria in the U.S. Still, congenital infections, infections acquired through<br />

exposure to infected blood or blood products, and infections acquired through local mosquitoborne<br />

transmission do occur. 2 In 2002, 1,337 cases of malaria, including eight deaths, were<br />

reported in the U.S. Other facts pertaining to malaria in the U.S. include: 3<br />

• Between 1957 and 2003, in the U.S., 63 outbreaks of locally transmitted mosquito-borne<br />

malaria have occurred. In such outbreaks, local mosquitoes become infected by biting<br />

persons carrying malaria parasites (acquired in endemic areas) and then transmit malaria<br />

to local residents.<br />

• Of the ten species of Anopheles mosquitoes found in the U.S., the two species that were<br />

responsible for malaria transmission prior to eradication (Anopheles quadrimaculatus in<br />

the east and Anopheles freeborni in the west) are still widely prevalent; thus there is a<br />

constant risk that malaria could be reintroduced in the U.S.<br />

• Malaria in humans is caused by one of four protozoan species of the genus Plasmodium:<br />

P. falciparum, P. vivax, P. ovale, or P. malariae. All are transmitted by the bite of an<br />

infected female Anopheles mosquito. 4<br />

• Of the 1,337 malaria cases reported for 2002 in the U.S., all but five were imported.<br />

Plasmodium falciparum, the most severe and life-threatening form of the disease, was<br />

identified in over 50% of the 1,337 cases in 2002.<br />

• During 1963-1999, 93 cases of transfusion-transmitted malaria were reported in the U.S.;<br />

approximately two thirds of these cases could have been prevented if the implicated<br />

donors had been deferred according to established guidelines.<br />

Antimalarial drugs are prescribed to treat infections from one of the four the Plasmodium species<br />

or to provide prophylactic protection to individuals who are visiting malaria-endemic countries<br />

from being infected. This review encompasses all dosage forms and strengths. Table 1 lists the<br />

single entity antimalarial drugs included in this review.<br />

Table 1. Single Entity Antimalarial Products in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name<br />

Chloroquine Phosphate Oral Tablet* / Injection Aralen Phosphate<br />

Chloroquine HCl Injection Aralen HCL<br />

Halofantrine HCl Not Available in US Halfan<br />

Hydroxychloroquine Sulfate Oral Tablet* Plaquenil<br />

Mefloquine HCl Oral Tablet* Lariam<br />

Primaquine Phosphate Oral Tablet* / Powder* Primaquine<br />

Pyrimethamine Oral Tablet Daraprim<br />

Quinine Sulfate Oral Capsule* /Oral Tablet* Quinine<br />

*Generic Available.<br />

275


II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Guidelines for the treatment and prevention of malaria have been established by the Centers for<br />

Disease Control and Prevention. The CDC guidelines provide clinicians clinical criteria to<br />

consider for appropriate selection of prophylactic therapy or treatment in malaria infected patients.<br />

Prophylaxis: 5<br />

Antimalarial agents prescribed for prophylactic purposes incorporate several strategies to prevent<br />

malarial infections. Primary prophylaxis is a strategy that uses antimalarial medications prior to,<br />

during, and after the exposure period to prevent the initial infection. The duration of therapy after<br />

the travel period is dependent on the antimalarial agent prescribed; four weeks after travel with<br />

chloroquine or mefloquine, seven days after travel with Atovaquone/proguanil. Drugs with longer<br />

half-lives, which are taken weekly, offer the advantage of a wider margin of error if the traveler is<br />

late with a dose than drugs with short half-lives, which are taken daily. Terminal prophylaxis is a<br />

strategy that uses an antimalarial drug towards the end of the exposure period, or immediately<br />

thereafter, to prevent relapses or delayed-onset clinical presentations of malaria caused by P. vivax<br />

or P. ovale. Primaquine is used in this purpose for terminal prophylaxis to decrease the risk of<br />

relapses by acting against the liver stages of the two plasmodium species. Primaquine is usually<br />

administered for 14 days after the traveler has left a malaria-endemic area. Terminal prophylaxis<br />

with primaquine for prevention of relapses is generally indicated for persons who have had<br />

prolonged exposure in malaria-endemic areas.<br />

In selecting an appropriate chemoprophylactic regimen before travel, the traveler and the healthcare<br />

provider should consider several factors. The travel itinerary should be reviewed and<br />

compared with the information on areas of risk in a given country to determine whether the<br />

traveler will actually be at risk for acquiring malaria. Whether the traveler will be at risk for<br />

acquiring drug-resistant P. falciparum malaria should also be determined.<br />

Resistance to antimalarial drugs has developed in many regions of the world. Chloroquineresistant<br />

P. falciparum has been confirmed in all areas with P. falciparum malaria except the<br />

Dominican Republic, Haiti, Central America west of the former Panama Canal Zone, Egypt, and<br />

some countries in the Middle East. In addition, resistance to sulfadoxine-pyrimethamine is<br />

widespread in the Amazon River Basin are of South America, much of Southeast Asia, other parts<br />

of Asia, and, increasingly, in large parts of Africa. Mefloquine resistance has been confirmed on<br />

the borders of Thailand with Burma and Cambodia, in the western provinces of Cambodia, and in<br />

the eastern states of Burma. Health-care providers are advised to consult with the latest<br />

information on resistance patterns before prescribing prophylaxis for their patients.<br />

Treatment: 6<br />

Treatment of malaria infections should not be initiated without confirmed laboratory diagnosis and<br />

should be initiated immediately thereafter. Three main factors should dictate the course of<br />

treatment in patients diagnosed with malaria:<br />

• The infecting Plasmodium species. P. falciparum infections can cause rapidly<br />

progressive, severe illness or death while the non-falciparum (P. vivax, P. ovale, or P.<br />

malariae) species rarely cause severe manifestations; P. vivax and P. ovale infections<br />

require treatment for the hypnozoite forms that remain dormant in the liver and can cause<br />

a relapsing infection; and P. falciparum and P. vivax species have different drug<br />

resistance patterns in differing geographic regions. For P. falciparum infections, the<br />

urgent initiation of appropriate therapy is especially critical.<br />

• The clinical status of the patient. Patients diagnosed with malaria are categorized as<br />

having either uncomplicated or severe malaria. Uncomplicated malaria can be effectively<br />

treated with oral antimalarial drugs. However, patients with one or more of the following<br />

symptoms are considered to have manifestations of more severe disease and should be<br />

treated aggressively with parenteral antimalarial therapy:<br />

276


• Impaired consciousness/coma<br />

• Severe normocytic anemia<br />

• Renal failure<br />

• Pulmonary edema<br />

• Acuter respiratory distress syndrome<br />

• Circulatory shock<br />

• Disseminated intravascular coagulation<br />

• Spontaneous bleeding<br />

• Acidosis<br />

• Hemoglobinuria<br />

• Jaundice<br />

• Repeated generalized convulsions<br />

• Parasitemia of > 5%<br />

• The drug susceptibility of the infecting parasites as determined by the geographic areas<br />

where the infection was acquired. Knowledge of the geographic area where the infection<br />

was acquired provides information on the likelihood of drug resistance of the infecting<br />

parasite and enables appropriate drug product, or drug combination selection and<br />

treatment course.<br />

All cases of malarial infections should be reported to state health departments which forward the<br />

information on to the CDC. Drug product selection and dosing in the treatment of malarial<br />

infections will be addressed further in this review. It is important to continue monitoring the<br />

patient’s clinical and parasitologic status after initiation of antimalarial therapy. Blood smears<br />

should be conducted to confirm adequate parasitologic response to treatment. In infections with P.<br />

falciparum or suspected chloroquine-resistant P. vivax, blood smears should show evidence of<br />

parasite density decreases, followed by clearance.<br />

277


Antimalarial Agents – Single Entity Products<br />

III.<br />

Comparative Indications for Single Entity Antimalarial Agents<br />

Table 2 lists the FDA-approved indications for single entity antimalarial agents.<br />

Table 2. FDA-Approved Indications for the Single-Entity Antimalarial Agents 7<br />

Prophylaxis<br />

Treatment<br />

Generic Drug Name P. vivax P. malariae P. ovale P. falciparum P. vivax P. malariae P. ovale P. falciparum<br />

Chloroquine Phosphate<br />

✔ ✔ ✔ ✔* ✔ ✔ ✔ ✔*<br />

Chloroquine HCl<br />

Hydroxychloroquine Sulfate<br />

Mefloquine HCl<br />

Primaquine Phosphate<br />

Pyrimethamine<br />

Quinine Sulfate<br />

✔ ✔ ✔ ✔*<br />

✔ ✔ ✔ ✔* ✔ ✔ ✔ ✔*<br />

✔ ✔** ✔ ✔**<br />

✔ *** ✔ *** ✔ *** ✔ ***<br />

✔<br />

✔ ✔ ✔ ✔**<br />

* Susceptible strains.<br />

**For susceptible strains of P. falciparum (both chloroquine-susceptible and resistant strains).<br />

***For chemoprophylaxis of susceptible strains of plasmodia only. Pyrimethamine is not suitable as a prophylactic agent for travelers to<br />

most areas because of prevalent resistance worldwide.<br />

Table 3. Malaria Prophylaxis and Treatment Therapy Choices<br />

Clinical Diagnosis or Plasmodium<br />

species<br />

Prophylaxis 8<br />

Treatment 9<br />

Areas where P. falciparum is sensitive to<br />

Chloroquine<br />

Areas where P. falciparum is resistant to<br />

Chloroquine<br />

Areas where P. falciparum is resistant to<br />

Chloroquine and Mefloquine<br />

Uncomplicated malaria / P. falciparum or<br />

species not identified.<br />

Drug(s) of Choice<br />

Chloroquine phosphate<br />

Alternative: Hydroxychloroquine sulfate.<br />

(Travelers unable to take Chloroquine or Hydroxychloroquine should take<br />

Mefloquine, Doxycycline, or Atovaquone/Proguanil)<br />

Mefloquine<br />

Atovaquone/Proguanil<br />

Doxycycline<br />

(Primaquine may be used in rare instances and after consultation with malaria<br />

experts and only in travelers unable to take Mefloquine, Atovaquone/Proguanil,<br />

or Doxycycline. Because Primaquine can cause fatal hemolysis in G6PDdeficient<br />

persons, laboratory testing is required to rule out G6PD deficiency.)<br />

Atovaquone/Proguanil<br />

Doxycycline<br />

Chloroquine-sensitive:<br />

Adult: Chloroquine phosphate<br />

Pediatric: Chloroquine phosphate<br />

Chloroquine-resistant or unknown resistance:<br />

Adult: (one of the following)<br />

1. Quinine sulfate plus one of the following:<br />

a. Doxycycline<br />

b. Tetracycline<br />

c. Clindamycin<br />

2. Mefloquine<br />

Pediatric: (one of the following)<br />

1. Quinine sulfate plus one of the following:<br />

a. Doxycycline<br />

b. Tetracycline<br />

c. Clindamycin<br />

2. Atovaquone/Proguanil<br />

278


Uncomplicated malaria / P. malariae<br />

Uncomplicated malaria / P. vivax or P.<br />

ovale<br />

Uncomplicated malaria / P. vivax in<br />

areas of Chloroquine resistance<br />

Uncomplicated malaria: alternatives for<br />

pregnant women<br />

Severe malaria / P. falciparum<br />

Adult: Chloroquine phosphate<br />

Pediatric: Chloroquine phosphate<br />

Adult: Chloroquine phosphate + Primaquine phosphate<br />

Pediatric: Chloroquine phosphate+ Primaquine phosphate<br />

Adult: (one of the following)<br />

1. Quinine sulfate + Primaquine phosphate + Doxycycline or<br />

Tetracycline<br />

2. Mefloquine plus Primaquine phosphate<br />

Pediatric: (one of the following)<br />

1. Quinine sulfate + Primaquine phosphate + Doxycycline or<br />

Tetracycline<br />

2. Mefloquine plus Primaquine phosphate<br />

Chloroquine-sensitive:<br />

Chloroquine phosphate<br />

Chloroquine-resistant P. falciparum:<br />

Quinine sulfate + Clindamycin<br />

Chloroquine-resistant P. vivax:<br />

Quinine sulfate<br />

Adult: Quinidine gluconate IV, plus one of the following:<br />

1. Doxycycline IV<br />

2. Tetracycline oral<br />

3. Clindamycin IV<br />

Pediatric: Quinidine gluconate IV, plus one of the following:<br />

1. Doxycycline IV<br />

2. Tetracycline oral<br />

3. Clindamycin IV<br />

279


IV.<br />

Pharmacokinetic Parameters<br />

Table 4 lists the pharmacokinetic parameters and mechanisms of action of the Single-Entity<br />

Antimalarial products.<br />

10, 11, 12, 13, 14<br />

Table 4. Pharmacokinetic Parameters of the Single-Entity Antimalarial Agents<br />

Drug Mechanism of Bioavailability Protein Metabolism Active<br />

Chloroquine<br />

Phosphate<br />

Chloroquine HCl<br />

Hydroxychloroquine<br />

Sulfate<br />

Mefloquine HCl<br />

Action<br />

Acts primarily<br />

as a blood<br />

schizonticide.<br />

The drug’s<br />

Antimalarial<br />

action is unclear<br />

Acts primarily<br />

as a blood<br />

schizonticide.<br />

The drug’s<br />

Antimalarial<br />

action is unclear<br />

Acts primarily<br />

as a blood<br />

schizonticide.<br />

The drug’s<br />

Antimalarial<br />

action is unclear<br />

Acts primarily<br />

as a blood<br />

schizonticide.<br />

The drug’s<br />

Antimalarial<br />

action is unclear<br />

Readily<br />

absorbed<br />

Readily<br />

absorbed<br />

Readily<br />

absorbed<br />

Absolute oral<br />

absorption has<br />

not been<br />

determined<br />

since an IV<br />

formulation is<br />

not available.<br />

The<br />

bioavailability<br />

of the tablet<br />

formulation<br />

compared with<br />

an oral solution<br />

was over 85%.<br />

Food<br />

significantly<br />

enhances the<br />

rate and extent<br />

of absorption<br />

by 40%.<br />

Binding<br />

55% Undergoes<br />

appreciable<br />

degradation in the<br />

tissues it is<br />

deposited. Up to<br />

70% of drug is<br />

excreted<br />

unchanged.<br />

55% Undergoes<br />

appreciable<br />

degradation in the<br />

tissues it is<br />

deposited. Up to<br />

70% of drug is<br />

excreted<br />

unchanged.<br />

55% Undergoes<br />

appreciable<br />

degradation in the<br />

tissues it is<br />

deposited. Up to<br />

70% of drug is<br />

excreted<br />

unchanged.<br />

Metabolites<br />

Yes<br />

Desethylchoroquine<br />

accounts for<br />

20% of urinary<br />

excretion<br />

Yes<br />

Desethylchoroquine<br />

accounts for<br />

20% of urinary<br />

excretion<br />

Yes<br />

Desethylchoroquine<br />

accounts for<br />

20% of urinary<br />

excretion<br />

Elimination<br />

Renal;<br />

Urinary<br />

excretion is<br />

enhanced by<br />

urinary<br />

acidification.<br />

Renal;<br />

Urinary<br />

excretion is<br />

enhanced by<br />

urinary<br />

acidification.<br />

Renal;<br />

Urinary<br />

excretion is<br />

enhanced by<br />

urinary<br />

acidification.<br />

98% Liver No Bile and<br />

Feces; 9% is<br />

excreted<br />

unchanged in<br />

urine<br />

Half-<br />

Life<br />

30-60<br />

days-<br />

30-60<br />

days<br />

30-60<br />

days<br />

2-4<br />

weeks


Primaquine<br />

Phosphate<br />

Pyrimethamine<br />

Quinine Sulfate<br />

Acts as a blood<br />

and tissue<br />

schizonticide<br />

that disrupts the<br />

parasite’s<br />

mitochondria<br />

and bind to<br />

native DNA.<br />

This activity<br />

disrupts the<br />

metabolic<br />

process and<br />

inhibits the<br />

gametocyte and<br />

exoerythhrocyte<br />

forms.<br />

A folic acid<br />

antagonist that<br />

possesses blood<br />

and some tissue<br />

schizonticidal<br />

activity.<br />

Acts primarily<br />

as a blood<br />

schizonticide.<br />

The drug’s<br />

Antimalarial<br />

action is unclear.<br />

Readily<br />

absorbed<br />

Readily<br />

absorbed<br />

Readily<br />

absorbed,<br />

mainly from<br />

the upper small<br />

intestine.<br />

98% Liver Yes<br />

Carboxyprimaquine<br />

with less<br />

antimalarial<br />

activity than<br />

parent drug.<br />

87% Liver Insufficient<br />

Data<br />

Feces; < 1%<br />

excreted<br />

unchanged in<br />

the urine.<br />

Renal.<br />

70-85% Liver No Renal;<br />

excretion is<br />

more rapid in<br />

acidic urine<br />

3-7<br />

hours<br />

4 days<br />

4-5<br />

hours<br />

V. Drug Interactions<br />

There are several drug interactions associated with Antimalarial agents that are addressed in Table<br />

5. Severity level 1 interactions were reported with quinine and mefloquine.<br />

Table 5. Drug Interactions of the Single-Entity Antimalarial Agents 15<br />

Drug Significance Interaction Mechanism<br />

Quinine Level 1 (delayed, major,<br />

suspected)<br />

Quinine and nonsedating<br />

antihistamines<br />

Quinine may inhibit the hepatic metabolism of<br />

Astemizole, increasing Astemizole concentrations<br />

Quinine<br />

Mefloquine<br />

Mefloquine<br />

Quinine<br />

Level 1 (delayed, major,<br />

suspected)<br />

Level 1 (rapid, major,<br />

suspected)<br />

Level 1 (delayed, major,<br />

suspected)<br />

Level 2 (rapid,<br />

moderate, suspected)<br />

Quinine and warfarin<br />

Halofantrine and Mefloquine<br />

Ziprasidone and Mefloquine<br />

Quinine and nondepolarizing muscle<br />

relaxants<br />

and possible cardiotoxicity.<br />

Quinine may inhibit the hepatically<br />

synthesized clotting factors and potentiate<br />

anticoagulation.<br />

Administration of halofantrine simultaneously or<br />

subsequent to Mefloquine can increase risk of lifethreatening<br />

arrhythmias through additive<br />

prolongation of the QT interval.<br />

Ziprasidone when administered in patients on<br />

Mefloquine can increase risk of life-threatening<br />

cardiac arrhythmias, including torsades de pointes<br />

through synergistic or additive prolongation of QT<br />

interval.<br />

The pharmacologic effects of nondepolarizing<br />

muscle relaxants may be enhanced by Quinine,<br />

requiring close monitoring of neuromuscular<br />

function.<br />

Quinine Level 2 (delayed,<br />

moderate, probable)<br />

Quinine and Digoxin<br />

Quinine appears to decrease the biliary clearance of<br />

Digoxin leading to a probable increase in serum<br />

Digoxin levels with possible toxicity.<br />

Quinine Level 2 (delayed, Ketoconazole and Quinine Ketoconazole inhibits Quinine metabolism<br />

281


Quinine<br />

Quinine<br />

Chloroquine<br />

Chloroquine<br />

Hydroxychloro<br />

quine<br />

Hydroxychloro<br />

quine<br />

Hydroxychloro<br />

quine<br />

Quinine<br />

Quinine<br />

Quinine<br />

Mefloquine<br />

Chloroquine<br />

Chloroquine<br />

Chloroquine<br />

Chloroquine<br />

Quinine<br />

moderate, suspected)<br />

Level 2 (delayed,<br />

Moderate, probable)<br />

Level 2 (rapid,<br />

moderate, suspected)<br />

Level 3 (delayed, minor,<br />

suspected)<br />

Level 3 (delayed, minor,<br />

suspected)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 4 (delayed, major,<br />

possible)<br />

Level 4 (rapid,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 4 (rapid,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Level 5 (delayed, minor,<br />

possible)<br />

Level 5 (delayed, minor,<br />

possible)<br />

Level 5 (delayed, minor,<br />

possible)<br />

Rifamycins and Quinine<br />

Quinine and Succinylcholine<br />

Cimetidine and Chloroquine<br />

Oral Magnesium Salts and<br />

Chloroquine<br />

Hydroxychloroquine and Betablockers<br />

Hydroxychloroquine and Betablockers<br />

Hydroxychloroquine and Digoxin<br />

Quinine and Amantadine<br />

Quinine and Carbamazepine<br />

Quinine and Phenobarbital<br />

Metoclopramide and Mefloquine<br />

Chloroquine and Cyclosporine<br />

Chloroquine and Penicillamine<br />

Chloroquine and Methotrexate<br />

Aluminum salts and Chloroquine<br />

Cimetidine and Quinine<br />

(CYP3A4). Quinine serum levels may be elevated,<br />

increasing therapeutic and toxic effects.<br />

Rifamycins increase the hepatic clearance of<br />

Quinine resulting in a reduction in Quinine’s<br />

therapeutic effects.<br />

Quinine may slow the metabolic rate for<br />

succinylcholine, thereby prolonging neuromuscular<br />

blockade produced by succinylcholine.<br />

The pharmacologic effects of chloroquine may be<br />

increased as a result of decreased Chloroquine<br />

metabolism caused by Cimetidine’s inhibition of<br />

hepatic mixed function oxidases.<br />

Oral Magnesium salts administered with<br />

Chloroquine may adsorb Chloroquine, decreasing<br />

Chloroquine’s therapeutic effect. The antacid<br />

activity of Magnesium salts may also be reduced.<br />

Plasma concentrations and cardiovascular effects of<br />

certain Beta-blockers may be increased due to<br />

Hydroxychloroquine’s inhibition of CYP2D6-<br />

mediated Beta-blocker metabolism.<br />

Hydroxychloroquine inhibits CYP2D6-mediated<br />

Beta-blocker metabolism, increasing plasma<br />

concentrations and CV effects of certain Betablockers.<br />

Mechanism unknown. Serum levels of Digoxin<br />

may increase, enhancing Digoxin action or toxicity.<br />

Quinine may inhibit renal clearance of Amantadine<br />

in men, causing elevations in serum Amantadine<br />

concentrations and increasing risk of toxicity.<br />

Quinine may inhibit Carbamazepine metabolism<br />

(CYP3A4), elevating Carbamazepine<br />

concentrations and increasing the pharmacologic<br />

and adverse effects.<br />

Quinine increases Phenobarbital serum<br />

concentrations, increasing the pharmacologic and<br />

adverse effects due to inhibition of Phenobarbital<br />

metabolism.<br />

Mefloquine’s serum concentrations may be elevated<br />

with possible increases in toxicity (GI, CNS, CV)<br />

due to metoclopramide’s effect in increasing gastric<br />

emptying time and increasing the rate of<br />

Mefloquine absorption in the small intestine.<br />

Chloroquine decreases Cyclosporin metabolism,<br />

elevating serum concentrations and increasing<br />

toxicity risks.<br />

Mechanism unknown. The pharmacologic and toxic<br />

effects of Penicillamine may be increased.<br />

Mechanism unknown. Chloroquine may reduce the<br />

bioavailability of Methotrexate, decreasing its<br />

antirheumatic effect.<br />

Oral Aluminum salts may adsorb Chloroquine,<br />

decreasing Chloroquine’s absorption and<br />

therapeutic effect.<br />

Cimetidine may reduce Quinine metabolism by<br />

inhibiting the hepatic microsomal mixed-function<br />

oxidase enzyme system, reducing Quinine clearance<br />

and increasing elimination half-life.<br />

282


Other interactions (per manufacturers labeling): 16<br />

• Quinine and Aluminum containing antacids: Aluminum-containing antacids may delay or<br />

decrease absorption of concurrent quinine.<br />

• Quinine and Mefloquine: Do not use concurrently. ECG abnormalities or cardiac arrest<br />

may occur as well as the risk of convulsions.<br />

• Quinine and Urinary alkalinizers: Urinary alkalinizers administered concurrently with<br />

quinine may increase quinine levels with potential toxicity.<br />

• Mefloquine and Beta-blockers: There is one reports of cardiopulmonary arrest with full<br />

recovery in a patient taking propranolol and mefloquine.<br />

• Mefloquine and Chloroquine: Concomitant use of mefloquine with chloroquine increases<br />

the risk of convulsion.<br />

• Mefloquine and Live attenuated bacterial vaccines: Vaccinations with attenuated live<br />

bacteria should be completed at least three days before the first dose of mefloquine.<br />

• Mefloquine and Anticonvulsants: Coadministration may reduce seizure control by<br />

lowering the plasma levels of the anticonvulsants.<br />

• Primaquine and Quinacrine: Do not administer Primaquine to patients who have recently<br />

received quinacrine due to the potential toxicity that may arise from antimalarial<br />

compounds which are structurally related to Primaquine.<br />

• Pyrimethamine and Antifolate drugs or agents associated with myelosuppression (e.g.<br />

Proguanil, zidovudine, methotrexate, sulfonamides, TMP-SMX): Concurrent use of<br />

antifolic acids and pyrimethamine may increase the risk of bone marrow suppression.<br />

• Pyrimethamine and Lorazepam: Mild hepatotoxicity has been reported when lorazepam<br />

and pyrimethamine were coadministered.<br />

VI.<br />

Adverse Drug Events of the Single-Entity Antimalarial Agents<br />

The majority of adverse drug reactions associated with the single-entity Antimalarial products tend<br />

to involve gastrointestinal disturbances or CNS effects. When used to treat for acute malaria,<br />

many symptoms possibly attributable to the drugs could not be distinguished from symptoms<br />

usually attributable to the disease itself.<br />

The 4-Aminoquinolone products (eg. Chloroquine, Hydroxychloroquine, Primaquine) should be<br />

used in extreme caution in patients with glucose-6-phosphate dehydrogenase deficiency.<br />

Table 6 lists the most common adverse events reported for these single-entity agents:<br />

17, 18, 19, 20, 21<br />

Table 6. Common Adverse Events (%) Reported for the Single-Entity Antimalarial Agents<br />

Adverse Event<br />

Body as a Whole<br />

Malaise<br />

Myalgia<br />

Cardiovascular<br />

Anginal Symptoms<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Vasculitis<br />

Bardycardia<br />

Tachycardia<br />

Extrasystoles<br />

Palpitations<br />

Chest Pain<br />

ECG Changes<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Hepatitis<br />

Appetite decrease<br />

Chloroquine<br />

Phosphate<br />

Chloroquine<br />

HCL<br />

Hydroxychloroquine<br />

Sulfate<br />

Mefloquine Primaquine Pyrimethamine Quinine<br />

- - - -<br />

- -<br />

b<br />

b<br />

-<br />

- -<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />


Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Syncope<br />

Restlessness<br />

Tinnitus<br />

Confusion<br />

Drowsiness<br />

Convulsions<br />

Emotional Disturbances<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Pruritus<br />

Urticaria<br />

Edema<br />

Pigment changes<br />

Hematologic<br />

Neutropenia<br />

Hemolytic anemia<br />

Agranulocytosis<br />

Thrombocytopenia<br />

Hypothrombinemia<br />

Granulocytopenia<br />

Leukopenia<br />

Blood dyscrasias<br />

Megaloblastic anemia<br />

Pancytopenia<br />

Hematuria<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

Ophthalmic<br />

Visual disturbances<br />

Photophobia<br />

Diplopia<br />

Mydriasis<br />

Optic atrophy<br />

Night blindness<br />

Retinal damage<br />

Other<br />

Cinchonism<br />

Hypoglycemia<br />

Psychotic/Paranoid Rx<br />

Cardiomyopathy<br />

Atrophic glossitis<br />

Rhythm disorders<br />

Pulmonary eosinophilia<br />

Hypersenstivity Rxn<br />

Hyperphenylalaninemia<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

-<br />

b<br />

b<br />

b<br />

b<br />

b<br />


VII.<br />

Dosing and Administration for the Single-Entity Antimalarial Agents<br />

22, 23, 24, 25, 26, 27<br />

Table 7. Dosing for the Single-Entity Antimalarial Agents<br />

Drug Availability Dose /Frequency/Duration<br />

Chloroquine<br />

Phosphate<br />

250mg tablet<br />

(Equiv to 150mg base)<br />

500mg tablet<br />

(Equiv to 300mg base)<br />

5mg injection<br />

(Equiv to 200mg base)<br />

Dosage of Chloroquine phosphate is often expressed in terms of equivalent chloroquine<br />

base.<br />

Malaria Suppression<br />

Adults – 300mg (base) weekly, on the same day each week. Begin 1-2 weeks prior to<br />

exposure; continue for 8 weeks after leaving endemic area<br />

If suppressive therapy is not begun prior to exposure, double the initial loading dose<br />

and give in two divided doses, 6 hours apart (adults – 600mg base; children – 10mg<br />

base/kg).<br />

Children – Administer 5mg base/kg weekly up to a maximum adult dose of 300mg<br />

base.<br />

Treatment of Acute Malaria<br />

Adult – An initial dose of 1gm (=600mg base) followed by an additional 500mg<br />

(=300mg base) after 6-8 hours and a single dose of 500mg (=300mg base) on each of<br />

two consecutive days. This represents a total dose of 2.5gm Chloroquine phosphate or<br />

1.5gm base in three days.<br />

Chloroquine HCl<br />

Hydroxychloroquine<br />

Sulfate<br />

50mg/ml injection<br />

(Equiv to 40mg/ml base)<br />

200mg tablet<br />

(Equiv to 155mg base)<br />

Adults of low body weight/infants/children – An initial dose of 10mg base per kg (but<br />

not exceeding a single dose of 600mg base). The second dose is administered 6 hours<br />

after the first dose and is 5mg base per kg (but should not exceed a single dose of<br />

300mg base). The third dose is administered 24 hours after the first dose and is 5mg<br />

base per kg. The fourth dose is administered 36 hours after the first dose and is 5mg<br />

base per kg.<br />

Treatment of Acute Malaria<br />

Adult – 160 to 200mg base (4-5ml) IM initially; repeat in 6 hours if necessary. Do not<br />

exceed 800mg (base) total dose in the first 24 hours. Begin oral dosage as soon as<br />

possible and continue for 3 days until approximately 1.5gm base has been<br />

administered.<br />

Other suggested dosages include 2.5mg base/kg every 4 hours or 3.5mg/kg every 6<br />

hours; repeat if necessary, maximum 25mg/kg/day.<br />

Children – Infants and children are extremely susceptible to overdosage. Severe<br />

reactions and deaths have occurred. The recommended single dose if 5mg base/kg;<br />

repeat in 6 hours. Do not exceed 10mg/kg/24 hours.<br />

Malaria Suppression<br />

Adults – 310mg (base) weekly, in the same day each week. Begin 1-2 weeks prior to<br />

exposure; continue for 8 weeks after leaving endemic area<br />

If suppressive therapy is not begun prior to exposure, double the initial loading dose<br />

and give in two divided doses, 6 hours apart (adults – 620mg base; children – 10mg<br />

base/kg).<br />

Children – Administer 5mg base/kg weekly up to a maximum adult dose.<br />

Treatment of Acute Malaria<br />

Adult – An initial dose of 620mg base followed by a second dose of 310mg base after<br />

6 hours and single doses of 310mg base on each of two consecutive days. This<br />

represents a total dose of 1.55gm base in three days. An alternative method employing<br />

a single dose of 620mg base, has also proved effective.<br />

Adults of low body weight/infants/children – An initial dose of 10mg base per kg (but<br />

not exceeding a single dose of 620mg base). The second dose is administered 6 hours<br />

285


after the first dose and is 5mg base per kg (but should not exceed a single dose of<br />

310mg base). The third dose is administered 18 hours after the second dose and is 5mg<br />

base per kg. The fourth dose is administered 24 hours after the third dose and is 5mg<br />

base per kg. This represents a total administered dose of 25mg base per kg of body<br />

weight over a three day period.<br />

Mefloquine 250mg tablet Malaria Suppression<br />

Adults – 250mg weekly, in the same day each week. Begin 1 week prior to exposure;<br />

continue for 4 weeks after leaving endemic area. Tablets should not be taken on an<br />

empty stomach and should be administered with at least 8oz of water.<br />

In certain cases (eg, when a traveler is taking other medication), it may be desirable to<br />

start prophylaxis 2-3 weeks prior to exposure, in order to ensure that the combination<br />

of drugs is well tolerated.<br />

Children – The recommended prophylactic dose of Mefloquine is approximately<br />

5mg/kg body weight once weekly. One 250mg Mefloquine tablet should taken once<br />

weekly in pediatric patients weighing over 45kg (100lbs). In pediatric patients<br />

weighing less than 45kg, the weekly dose decreases in proportion to body weight:<br />

30 to 45kg: ¾ tablet<br />

20 to 30kg ½ tablet<br />

10 to 20kg ¼ tablet<br />

5 to 10kg 1/8 tablet<br />

Experience with Mefloquine in infants less than 3 months old or weighing less than<br />

5kg is limited.<br />

Treatment of Mild to Moderate Malaria<br />

Adults caused by P. vivax or Mefloquine-susceptible strains of P. Falciparum – Five<br />

tablets (1,250mg) Mefloquine to be given as a single oral dose. The drug should not be<br />

taken on an empty stomach and should be administered with at least 8oz of water.<br />

Children caused by Mefloquine-susceptible strains of P. Falciparum – 20 to 25mg/kg<br />

body weight. Splitting the total therapeutic dose into 2 doses taken 6-8 hours apart may<br />

reduce the occurrence or severity of adverse effects. Experience with Mefloquine in<br />

infants less than 3 months old or weighing less than 5kg is limited. The drug should<br />

not be taken on an empty stomach and should be administered with ample water. The<br />

tablets may be crushed and suspended in a small amount of water, milk or other<br />

beverage for administration to small children and other persons unable to swallow them<br />

whole.<br />

In pediatric patients, the administration of Mefloquine for the treatment of malaria has<br />

been associated with early vomiting. In some cases, early vomiting has been cited as a<br />

possible cause of treatment failure. If a significant loss of drug product is observed or<br />

suspected because of vomiting, a second full dose should be administered to patients<br />

who vomit less than 30 minutes after receiving the drug. If vomiting occurs 30-60<br />

minutes after a dose, an additional half-dose should be given. If vomiting occurs, the<br />

patient should be monitored closely and alternative malaria treatment considered if<br />

improvement is not observed within a reasonable period of time.<br />

The safety and effectiveness of Mefloquine to treat malaria in children below the age of<br />

6 months have not been established.<br />

Primaquine<br />

26.3mg tablet<br />

(Equiv to 15mg base)<br />

In both adults and children, if a full-treatment course with Mefloquine does not lead to<br />

improvement within 48 to 72 hours, Mefloquine should not be used for retreatment.<br />

An alternative therapy should be used. Similarly, if previous prophylaxis with<br />

Mefloquine has failed, Mefloquine should not be used for curative treatment.<br />

Treatment of P. vivax Malaria<br />

Patients suffering from an attack of P. vivax malaria or having parasitized red blood<br />

cells should receive a course of Chloroquine.<br />

Adult – 30mg (base) taken orally daily for 14 days.<br />

Children – 0.6mg base/kg taken orally, daily for 14 days.<br />

286


Pyrimethamine 25mg tablet Malaria Suppression<br />

Pyrimethamine is rapidly absorbed and therefore prophylactic cover can be expected<br />

shortly after the first dose. Prophylaxis should commence before arrival in an endemic<br />

area and be continued for four weeks after returning to a non-malarious area.<br />

Adults and children >10 years of age – 25mg orally every week.<br />

Children (4-10 years of age) – 12.5mg orally every week.<br />

Infants and children 10 years of age – 50mg orally daily for 2 days.<br />

Children (4-10 years of age) – 25mg orally daily for 2 days.<br />

Infants and children


Hydroxychloroquine<br />

Sulfate<br />

No No No special dosing consideration NR Crushing tablets and putting each dose in<br />

capsules; contents of capsules may be<br />

mixed with jam, jelly, or jello.<br />

Mefloquine No No WHO recommends mefloquine<br />

for all children and infants 3<br />

months or older, or 5 kilograms<br />

or more in areas where<br />

chloroquine resistance is<br />

prevalent<br />

C<br />

Crushing tablet and suspending in water,<br />

milk, or other beverage if patient is<br />

unable to swallow it whole.<br />

Primaquine No No No special dosing consideration NR No data is available on crushing of tablet.<br />

Pyrimethamine No No No special dosing consideration C Tablets may be split. The manufacturer<br />

reports no data is available on the<br />

bioequivalency of crushing the tablets for<br />

use in suspensions, nor is data available<br />

on use of the suspension when given per<br />

tube.<br />

Quinine No Special<br />

caution in<br />

the use of<br />

Quinine in<br />

patients with<br />

concurrent<br />

liver<br />

disease.<br />

No special dosing consideration X No data is available on crushing of tablet<br />

or emptying contents of capsule.<br />

288


VIII. Comparative Effectiveness of the Single-Entity Antimalarial Agents<br />

Not all of the agents in this class can be used for the same malarial infections.<br />

Table 9. Additional Outcomes Evidence for the Single-Entity Antimalarial Agents<br />

Study Sample Treatment /<br />

Results<br />

Duration<br />

Ibrahim<br />

MH, et<br />

al 31 n=98 Quinine PO/IV In comparing 3 quinine treatment regimens against P. Falciparum malaria that had<br />

failed to respond to chloroquine treatment, the 93 patients were randomly allocated to<br />

1 of 3 regimens for seven days of therapy:<br />

• PO/IV quinine at doses 10mg/kg TID (32 patients)<br />

• PO/IV quinine at doses 10mg/kg BID (31 patients)<br />

• PO/IV quinine at doses 15mg/kg QD<br />

There was no significant difference in the parasite-clearance times observed in the<br />

three groups. In the treatment of chloroquine-resistant P. falciparum malaria in<br />

Sudan, once-daily treatment with quinine, in a relatively low daily dose, appears as<br />

effective as the TID treatment often recommended.<br />

White NJ,<br />

et al 32 n=50 Chloroquine IM<br />

3.5mg (base)/kg q6h<br />

An open paired randomized comparison of chloroquine IM and quinine IM was<br />

conducted in 50 Gambian children with severe falciparum malaria:<br />

Quinine IM 20mg<br />

(salt)/kg, followed by<br />

• 8 children died, 6 from the quinine-treated, and 2 from the chloroquinetreated.<br />

10mg/kg q12h. • Chloroquine reduced parasitaemia significantly more rapidly than did<br />

quinine, but other measures of the therapeutic response were similar in the<br />

two groups.<br />

• The findings did not support the proposition that quinine is intrinsically<br />

superior to chloroquine in the treatment of severe drug-sensitive falciparum<br />

Baird JK,<br />

et al 33 n=99 Primaquine 0.5mg<br />

base/kg QOD<br />

Chloroquine 5mg<br />

base/kg Qwk<br />

Mengesha Metaanalysis<br />

T, et al 34<br />

Chloroquine<br />

Amodiaquine<br />

Mefloquine<br />

Sulfadoxine-<br />

Pyrimethamine<br />

Halofantrine<br />

malaria.<br />

A controlled clinical trial comparing primaquine versus chloroquine for prophylaxis<br />

among non-immune transmigrants for Java and Bali in the region of Irian Jay,<br />

Indonesia. 45 subjects received 0.5mg of primaquine base/kg QOD, and 54 people in<br />

the same village received weekly 5mg of chloroquine base/kg for 16-19 weeks.<br />

The subjects taking chloroquine were 5 times more likely to suffer an episode of<br />

malaria than those taking primaquine. The minimum protective efficacy of<br />

primaquine was 74% for P. falciparum and 90% for P vivax infections.<br />

A meta-analysis study evaluated the efficacy and safety of chloroquine and alternative<br />

Antimalarial drugs used in six African countries including Ethiopia, Kenya, Uganda,<br />

Cote D’Ivoire, Gambia and Nigeria. Findings from the 6 countries showed a higher<br />

efficacy of amodiaquine and quinine >90%) in malaria treatment compared to<br />

chloroquine (>70%).<br />

Pharmacokinetic profile demonstrates that all these drugs have similar therapeutic<br />

effects, but differ in their adverse reactions, contraindications, and half-life.<br />

Randriana<br />

rivelojosia<br />

M, et al 35 - - Plasmodium falciparum isolates in patients with uncomplicated malaria attack were<br />

used in 293 in-vitro tests conducted with at least one Antimalarial agent. The purpose<br />

of the in-vitro testing was to determine the sensitivity of Plasmodium falciparum to<br />

the major Antimalarial drugs in Madagascar:<br />

• All of the successfully tested isolates were sensitive to halofantrine (n=56).<br />

• All of the successfully tested isolates were sensitive to quinine (n=199).<br />

• 94.2% of the successfully tested isolates were sensitive to chloroquine<br />

(n=205).<br />

• 98% of the successfully tested isolates were sensitive to mefloquine<br />

(n=199)<br />

The in-vitro sensitivity of P. falciparum to quinine, mefloquine and halofantrine<br />

encourages the use of these drugs as alternative in case of chloroquine treatment<br />

failures.


Schwartz<br />

E, et al 36 n=106 Prophylactic<br />

Therapies involving:<br />

Primaquine<br />

Doxycycline<br />

Mefloquine<br />

Kofoed K,<br />

et al 37 n=320 Prophylactic<br />

Therapies involving:<br />

Chloroquine/proguanil<br />

Mefloquine<br />

Atovaquone/proguanil<br />

A retrospective analysis of travelers who joined rafting trips in an area in Ethiopia<br />

where both P. vivax and P. falciparum are hyperendemic:<br />

• Of the 106 travelers who received primaquine, 5.7% developed malaria.<br />

• Of the 19 doxycycline recipients, 53% developed malaria.<br />

• Of the 25 mefloquine recipients, 52% developed P. vivax malaria (> 3<br />

months after return from the area).<br />

Primaquine was shown to be a safe and effective prophylactic drug against both P.<br />

falciparum and P. vivax malaria in travelers.<br />

An open case-control study was initiated to study 320 permanent residents in<br />

Denmark who were returning from abroad with malaria. This group was compared<br />

with a group of 600 travelers who were not infected with malaria and matched by age,<br />

sex, and destination. Information on the use of chemoprophylaxis and the length of<br />

stay in malarious areas were obtained by questionnaire.<br />

200 cases of P falciparum malaria were identified of which 103 had used<br />

chloroquine/proguanil, 16 used mefloquine, and 3 used atovaquone/proguanil as<br />

prophylaxis; the others had taken other drugs or nor prophylaxis:<br />

• The risk of malaria increased with increasing exposure.<br />

• Compliance to prophylactic treatment was lower, especially for mefloquine<br />

users in malaria cases compared to controls.<br />

• The estimated efficacy of chloroquine/proguanil, mefloquine, and<br />

atovaquone/proguanil in fully compliant users was 1:599, 1:2,232 and<br />

1:1,943, respectively, P. falciparum cases per prescription.<br />

• Chloroquine/proguanil was less efficient compared with mefloquine.<br />

Warhurst<br />

DC,et al 38 - - An analysis conducted to determine whether patients treated for autoimmune disease<br />

with hydroxychloroquine could achieve effective prophylaxis on<br />

hydroxychloroquine/proguanil against chloroquine-resistant P. falciparum, in place of<br />

the traditional prophylactic regimen of adding chloroquine/proguanil and the likely<br />

toxicity that would arise when Chloroquine is administered in persons being treated<br />

with hydroxychloroquine.<br />

Weiss<br />

WR, et<br />

al 39 - Prophylactic regimens<br />

of:<br />

1. Daily Primaquine<br />

2. Daily Doxycycline<br />

3. Weekly Chloroquine<br />

with proguanil<br />

4. Weekly mefloquine<br />

with vitamin<br />

5. Daily vitamin<br />

The in-vitro test results confirmed that hydroxychloroquine/proguanil prophylaxis<br />

was 8.8 times less active in a chloroquine-resistant P. falciparum than<br />

chloroquine/proguanil treatment. Hydroxychloroquine is therefore contraindicated in<br />

prophylaxis of treatment of chloroquine-resistant falciparum malaria.<br />

Primaquine was tested as a prophylactic drug against P. falciparum in a region in<br />

western Kenya in which malaria his holoendemic. Children 9-14 years old were<br />

randomized to receive regimens of daily primaquine, daily doxycycline, daily<br />

proguanil plus weekly chloroquine, daily vitamin plus weekly mefloquine, or daily<br />

vitamin alone. Primaquine, doxycycline, and mefloquine were equally effective in<br />

preventing both symptomatic and asymptomatic malarial infections. Chloroquine<br />

plus proguanil was the least effective regimen. There was not toxicity from daily<br />

primaquine during the 11 weeks of the study. Findings show that primaquine can be<br />

successfully used as a causal prophylactic regimen against falciparum malaria in<br />

western Kenya; chloroquine plus proguanil was not as efficacious as the three other<br />

preventive regimens.<br />

Additional Evidence<br />

Dose Simplification: In most cases, the antimalarial agents in this class are given as a single dose<br />

or for a brief duration (acute use), depending on the malarial plasmodium being treated. When<br />

given for longer durations, most doses are prescribed once weekly, given on the same day of the<br />

week. Malaria treatment often requires initial dosing, followed by a second dose in the same day,<br />

and then subsequent daily dosing for 2-3 days. Mefloquine offers a single oral treatment dose,<br />

while quinine is dosed three times daily for up to 12 days. No data was found in a literature search<br />

looking at the dosing frequency patterns of the antimalarial drugs and any benefit of single oral<br />

treatment doses on the outcome of disease.<br />

290


Stable Therapy: Significant clinical data is available on resistance to the Antimalarial agents in<br />

humans. Guidelines established by the CDC consider resistance prevalence in relation to the<br />

endemic malarial regions and origins of infection. Chloroquine-resistant P. falciparum has been<br />

confirmed in all areas with P. falciparum malaria except the Dominican Republic, Haiti, Central<br />

America west of the former Panama Canal Zone, Egypt, and some countries in the Middle East.<br />

In addition, resistance to sulfadoxine/pyrimethamine is widespread in the Amazon River Basin<br />

area of South America, much of Southeast Asia, other parts of Asia, and increasingly in large parts<br />

of Africa. Resistance to Mefloquine has been confirmed on the borders of Thailand with Burma<br />

and Cambodia, in the western provinces of Cambodia, and in the eastern states of Burma<br />

Because of prevalent resistant plasmodium strains, and varying side effects among the antimalarial<br />

agents, health care providers need to closely monitor patient response to therapy regimens and<br />

modify, if necessary to ensure successful suppression or treatment.<br />

Impact on Physician Visits: A literature search did not reveal clinical literature relevant to use of<br />

the antimalarials and their impact on physician visits.<br />

IX.<br />

Conclusions<br />

Antimalarial agents recommended in the suppression and treatment of malaria by the CDC include<br />

chloroquine products, hydroxychloroquine, mefloquine, primaquine, quinine, and the combination<br />

agent, atovaquone-proguanil. All single-entity antimalarial products listed in the CDC guideline<br />

have generic equivalent products on the market.<br />

All brand products within the single-entity antimalarial agents are comparable to the generics in<br />

this class and offer no significant clinical advantage over other alternatives in general use.<br />

X. Recommendations<br />

No brand single entity antimalarial product is recommended for preferred status.<br />

291


Antimalarial Agents – Combination products<br />

This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this<br />

review.<br />

Table 1. Combination Antimalarial Products in the <strong>Review</strong><br />

Generic Name Formulation Example Brand Name<br />

Atovaquone and Proguanil HCl Oral Tablet Malarone<br />

Malarone Pediatric<br />

Pyrimethamine and Sulfadoxine Oral Tablet Fansidar<br />

I. Comparative Indications for Combination Antimalarial Agents<br />

Table 2 lists the FDA-approved indications for combination antimalarial agents.<br />

Table 2. FDA-Approved Indications for the Combination Antimalarial Agents 40<br />

Prophylaxis<br />

Treatment<br />

Generic Drug Name P. vivax P. malariae P. ovale P. falciparum P. vivax P. malariae P. ovale P. falciparum<br />

Sulfadoxine/Pyrimethamine<br />

✔** ✔*<br />

Atovaquone/Proguanil HCl<br />

* Susceptible strains.<br />

** Malaria prophylaxis with Sulfadoxine/Pyrimethamine is not routinely recommended and should only be considered to travelers to areas<br />

where chloroquine-resistant P. falciparum malaria is endemic and sensitive to Sulfadoxine/Pyrimethamine, and when alternative drugs are<br />

not available or are contraindicated. However, strains of P. falciparum may be encountered which have developed resistance to<br />

Sulfadoxine/Pyrimethamine.<br />

✔<br />

✔<br />

Table 3. Malaria Prophylaxis and Treatment Therapy Choices<br />

Clinical Diagnosis or Plasmodium<br />

species<br />

Prophylaxis 41<br />

Areas where P. falciparum is sensitive to<br />

Chloroquine<br />

Areas where P. falciparum is resistant to<br />

Chloroquine<br />

Areas where P. falciparum is resistant to<br />

Chloroquine and Mefloquine<br />

Drug(s) of Choice<br />

Chloroquine phosphate<br />

Alternative: Hydroxychloroquine sulfate.<br />

(Travelers unable to take Chloroquine or Hydroxychloroquine should take<br />

Mefloquine, Doxycycline, or Atovaquone/Proguanil)<br />

Mefloquine<br />

Atovaquone/Proguanil<br />

Doxycycline<br />

(Primaquine may be used in rare instances and after consultation with malaria<br />

experts and only in travelers unable to take Mefloquine, Atovaquone/Proguanil,<br />

or Doxycycline. Because Primaquine can cause fatal hemolysis in G6PDdeficient<br />

persons, laboratory testing is required to rule out G6PD deficiency.)<br />

Atovaquone/Proguanil<br />

Doxycycline<br />

292


Treatment 42<br />

Uncomplicated malaria / P. falciparum or<br />

species not identified.<br />

Chloroquine-sensitive:<br />

Adult: Chloroquine phosphate<br />

Pediatric: Chloroquine phosphate<br />

Chloroquine-resistant or unknown resistance:<br />

Adult: (one of the following)<br />

3. Quinine sulfate plus one of the following:<br />

a. Doxycycline<br />

b. Tetracycline<br />

c. Clindamycin<br />

4. Mefloquine<br />

Pediatric: (one of the following)<br />

3. Quinine sulfate plus one of the following:<br />

a. Doxycycline<br />

b. Tetracycline<br />

c. Clindamycin<br />

4. Atovaquone/Proguanil<br />

293


II.<br />

Pharmacokinetic Parameters<br />

Table 4 lists the pharmacokinetic parameters and mechanisms of action of the combination<br />

antimalarial products.<br />

43, 44, 45, 46<br />

Table 4. Pharmacokinetic Parameters of the Combination Antimalarial Agents<br />

Drug Mechanism of Action Bioavailability Protein Metabolism Active<br />

Sulfadoxine /<br />

Pyrimethamine<br />

Sulfadoxine –<br />

synergistically acts at the<br />

state immediately<br />

preceding that of<br />

pyrimethamine.<br />

Pyrimethamine –<br />

interferes with the<br />

synthesis of folinic acid<br />

within the parasite<br />

inhibiting the conversion<br />

of dihydrofolate to<br />

tetrahydrofolate.<br />

Both<br />

compounds are<br />

readily absorbed<br />

Binding<br />

Sulf =<br />

90-95%<br />

Pyrim =<br />

87%<br />

Both<br />

compounds<br />

are<br />

metabolized<br />

in the liver<br />

Elimination Half-<br />

Metabolites<br />

Life<br />

No Renal Sulf =<br />

200hrs<br />

Pyri =<br />

100hrs<br />

Atovaquone /<br />

Proguanil HCl<br />

Together, the combination<br />

achieves a sequential<br />

blockade of 2 enzymes<br />

involved in the<br />

biosynthesis of folinic<br />

acid within the parasite.<br />

Atovaquone – action has<br />

not been fully elucidated.<br />

It is thought that<br />

Atovaquone inhibits<br />

mitochondrial electron<br />

transport of P. falciparum<br />

and thus pyrimidine<br />

synthesis.<br />

Proguanil HCl – requires<br />

metabolism to cycloguanil<br />

for Antimalarial activity;<br />

cycloguanil is a potent<br />

inhibitor of plasmodial<br />

dihydrofolate Reductase,<br />

resulting in inhibition of<br />

nucleic acid synthesis<br />

Atovaquone is<br />

highly lipophilic<br />

with low<br />

aqueous<br />

solubility.<br />

Bioavailability<br />

of Atovaquone<br />

(23%) shows<br />

considerable<br />

variability from<br />

patient to<br />

patient.<br />

Proguanil is<br />

readily absorbed<br />

Atov =<br />

>99%<br />

Prog =<br />

75%<br />

Atovaquone<br />

is not<br />

metabolized<br />

Proguanil is<br />

extensively<br />

metabolized<br />

in the liver to<br />

the active<br />

metabolite.<br />

Yes<br />

Proguanil is a<br />

prodrug that is<br />

metabolized to<br />

the active<br />

metabolite,<br />

cycloguanil via<br />

hepatic<br />

cytochrome<br />

P450<br />

isoenzyme<br />

CYP2C19.<br />

Atovaquone<br />

is excreted<br />

in the feces<br />

unchanged.<br />

Proguanil is<br />

excreted<br />

renally as<br />

the active<br />

metabolite.<br />

Atov<br />

= 2-3<br />

days<br />

Prog =<br />

12-<br />

21hrs<br />

Together, the synergistic<br />

activity of the two agents<br />

in falciparum malaria is<br />

unclear; an enhanced<br />

effect of cycloguanil,<br />

facilitated by the<br />

Atovaquone mediated<br />

reduction in pyrimidine<br />

nucleotide pools has been<br />

speculated.<br />

294


III.<br />

Drug Interactions<br />

There are several drug interactions associated with Combination Antimalarial agents that are<br />

addressed in Table 5.<br />

Table 5. Drug Interactions of the Combination Antimalarial Agents 47<br />

Drug Significance Interaction Mechanism<br />

Atovaquone Level 2 (delayed,<br />

moderate, suspected)<br />

Atovaquone and Zidovudine<br />

Atovaquone appears to elevate serum Zidovudine<br />

concentrations by inhibiting the glucuronidation of<br />

Zidovudine, increasing the risk of Zidovudine<br />

Atovaquone<br />

Atovaquone<br />

Level 4 (delayed,<br />

moderate, Possible)<br />

Level 5 (delayed,<br />

moderate, unlikely)<br />

Atovaquone and Etoposide<br />

Atovaquone and Azithromycin<br />

toxicity.<br />

Mechanism unknown. Etoposide plasma<br />

concentration may be elevated, increasing risk<br />

of toxicity.<br />

Mechanism unknown. Azithromycin serum<br />

concentrations may be reduced, decreasing the<br />

pharmacologic effect.<br />

Other interactions (per manufacturers labeling): 48<br />

• Sulfadoxine/Pyrimethamine and Chloroquine: There have been reports that may indicate<br />

an increase in incidence and severity of adverse reactions when chloroquine is used with<br />

Sulfadoxine/Pyrimethamine tablets as compared with the use of<br />

Sulfadoxine/Pyrimethamine tablets alone.<br />

• Sulfadoxine/Pyrimethamine and Antifolic drugs: Do not use antifolic drugs (eg,<br />

sulfonamides or TMP-SMX combinations) while the patient is receiving<br />

Sulfadoxine/Pyrimethamine tablets for Antimalarial prophylaxis.<br />

• Sulfadoxine/Pyrimethamine and Goitrogens, diuretics, hypoglycemic agents: The<br />

sulfonamides bear certain chemical similarities to some goitrogens, diuretics<br />

(acetazolamide and the thiazides), and oral hypoglycemic agents. Diuresis and<br />

hypoglycemia have occurred rarely in patients receiving sulfonamides. Cross-sensitivity<br />

may exist with these agents.<br />

• Atovaquone/Proguanil and Tetracycline: Concomitant treatment with tetracyclines has<br />

been associated with approximately 40% reduction in plasma concentrations of<br />

Atovaquone.<br />

• Atovaquone/Proguanil and Metoclopramide: Concomitant treatment with<br />

metoclopramide has been associated with decreased bioavailability of Atovaquone.<br />

• Atovaquone/Proguanil and Rifampin: Concomitant administration of rifampin is known<br />

to reduce Atovaquone levels by approximately 50%. The concomitant administration of<br />

these agents is not recommended. The mechanism of this interaction is unknown.<br />

IV.<br />

Adverse Drug Events of the Combination Antimalarial Agents<br />

The majority of adverse drug reactions associated with the single-entity Antimalarial products tend<br />

to involve gastrointestinal disturbances or CNS effects. When used to treat for acute malaria,<br />

many symptoms possibly attributable to the drugs, could not be distinguished from symptoms<br />

usually attributable to the disease itself.<br />

The manufacturer of Sulfadoxine/Pyrimethamine has issued a warning of the fatalities that have<br />

occurred with their product due to severe reactions, including Stevens-Johnson syndrome and<br />

toxic epidermal necrolysis. Discontinuation of therapy is advised at the first appearance of skin<br />

rash, if a significant reduction in the count of any formed blood element is noted, or upon the<br />

occurrence of active bacterial or fungal infections. In addition, fatalities associated with the<br />

administration of sulfonamides have occurred due to severe reactions, including fulminant hepatic<br />

necrosis, agranulocytosis, aplastic anemia and other blood dyscrasias. While severe, these<br />

reactions are rare.<br />

295


Table 6. Common Adverse Events (%) Reported for the Combination Antimalarial<br />

49, 50, 51, 52<br />

Agents<br />

Adverse Event Sulfadoxine/Pyrimethamine Atovaquone/Proguanil HCl<br />

Body as a Whole<br />

Malaise<br />

Myalgia<br />

Chills<br />

-<br />

b<br />

b<br />

b<br />

-<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Appetite decrease<br />

Glossitis<br />

Stomatitis<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Syncope<br />

Restlessness/Insomnia<br />

Tinnitus<br />

Convulsions<br />

Mental Depression<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Hepatic Necrosis<br />

Hepatic Granulomas<br />

Skin and Appendages<br />

Dermatitis<br />

Rash<br />

Pruritus<br />

Urticaria<br />

Phototoxicity<br />

Pigment changes<br />

Toxic Epidermal Necrolysis<br />

Erythema Nodosum<br />

Hematologic<br />

Neutropenia<br />

Hemolytic anemia<br />

Agranulocytosis<br />

Thrombocytopenia<br />

Hypothrombinemia<br />

Granulocytopenia<br />

Leukopenia<br />

Megaloblastic anemia<br />

Pancytopenia<br />

Thombocytopenia Purpura<br />

General Anemia<br />

Renal<br />

Crystalluria<br />

Nephrotoxicity<br />

Ophthalmic<br />

Iritis<br />

Amblyopia<br />

Other<br />

Periarteritis Nodosa<br />

Apathy<br />

Bronchospasms<br />

Pulmonary Eosinophilia<br />

Stevens-Johnson Syndrome<br />

Spontaneous Abortion<br />

Hypoalbuminemia<br />

Pulmonary infiltrates<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

-<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

bAdverse event reported; specific percentages not available<br />

17%<br />

12%<br />

8%<br />

5%<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

b<br />

-<br />

b<br />

296


V. Dosing and Administration for the Combination Antimalarial Agents<br />

53, 54<br />

Table 7. Dosing for the Combination Antimalarial Agents<br />

Drug Availability Dose /Frequency/Duration<br />

Sulfadoxine<br />

/Pyrimethamine<br />

500mg Sulfadoxine /<br />

25mg Pyrimethamine<br />

Tablets<br />

Malaria Suppression<br />

The first dose of Sulfadoxine 500mg/Pyrimethamine 25mg is taken 1 or 2 days before<br />

departure to an endemic area; continue administration during the stay and for 4-6 weeks<br />

after return.<br />

Age Once Weekly Once Every 2 Weeks<br />

Adults<br />

Pediatric (>2mo to 18yrs)<br />

>45 kg<br />

31-45kg<br />

21-30kg<br />

11-20kg<br />

5-10kg<br />

1 Tablet<br />

1½ Tablet<br />

1 Tablet<br />

¾ Tablet<br />

½ Tablet<br />

¼ Tablet<br />

2 Tablets<br />

NA<br />

NA<br />

NA<br />

NA<br />

NA<br />

Treatment of Acute Malaria<br />

A single dose of the following number of Sulfadoxine 500mg/Pyrimethamine 25mg<br />

tablets is used in sequence with Quinine or alone:<br />

Atovaquone<br />

/Proguanil HCl<br />

250mg Atovaquone /<br />

100mg Proguanil HCl<br />

Tablet<br />

62.5mg Atovaquone /<br />

25mg Proguanil HCl<br />

Pediatric Tablet<br />

Age<br />

Single Dose<br />

Adults<br />

2-3 Tablets<br />

Pediatric (>2mo to 18yrs)<br />

>45 kg<br />

3 Tablets<br />

31-45kg<br />

2 Tablets<br />

21-30kg<br />

1½ Tablets<br />

11-20kg<br />

1 Tablet<br />

5-10kg<br />

½ Tablet<br />

Malaria Suppression<br />

Start prophylactic treatment with Atovaquone/Proguanil 1 or 2 days before entering a<br />

malaria-endemic area and continue daily during the stay and for 7 days after return.<br />

Adults – Take 250mg Atovaquone/100mg Proguanil HCl (1 tablet) orally per day.<br />

Children – The dosage for prevention of malaria in pediatric patients is based upon body<br />

weight:<br />

Weight<br />

(kg)<br />

Atovaquone/Proguanil<br />

Total Daily Dose<br />

Dosage Regimen<br />

11-20 62.5mg/25mg 1 pediatric tablet daily<br />

21-30 125mg/50mg 2 pediatric tablets as a single daily dose<br />

31-40 187.5mg/75mg 3 pediatric tablets as a single daily dose<br />

> 40 250mg/100mg 1 adult strength tablet as a single daily dose<br />

Treatment of Acute Malaria<br />

Adults – Take 1000mg Atovaquone/400mg Proguanil HCl (4 tablets) orally as a single<br />

daily dose for 3 consecutive days.<br />

Children – The dosage for treatment of malaria in pediatric patients is based upon body<br />

weight:<br />

Weight<br />

(kg)<br />

Atovaquone/Proguanil<br />

Total Daily Dose<br />

Dosage Regimen<br />

5-8 125mg/50mg 2 pediatric tablets daily for 3 consecutive days<br />

9-10 187.5mg/75mg 3 pediatric tablets daily for 3 consecutive days<br />

11-20 250mg/100mg 1 adult strength tablet daily for 3 consecutive<br />

days<br />

21-30 500mg/200mg 2 adult strength tablet daily for 3 consecutive<br />

days<br />

31-40 750mg/300mg 3 adult strength tablet daily for 3 consecutive<br />

days<br />

> 40 1000mg/400mg 4 adult strength tablet daily for 3 consecutive<br />

days<br />

297


Special Dosing Considerations<br />

55, 56, 57<br />

Table 8. Special Dosing Considerations for the Combination Antimalarial Agents<br />

Drug Renal Dosing Hepatic Pediatric Use Pregnancy<br />

Sulfadoxine<br />

/Pyrimethamine<br />

Atovaquone<br />

/Proguanil HCl<br />

Dosing<br />

No No Sulfadoxine/Pyrimethamine<br />

should not be given to infants<br />

less than 2 months of age<br />

because of inadequate<br />

development of the glucuronideforming<br />

enzyme system.<br />

Yes<br />

Should not be<br />

used in patients<br />

with severe renal<br />

impairment<br />

(CrCl


Bustos DG, et<br />

al 60 n=110 Malaria treatment regimens<br />

of:<br />

1. Atovaquone/prog<br />

uanil<br />

2. Chloroquine<br />

3. Chloroquine,<br />

sulfadoxine,<br />

pyrimethamine<br />

Results:<br />

• Cure rates did not differ significantly between patients treated with<br />

atovaquone/proguanil (100%) and those treated with<br />

pyrimethamine/sulfadoxine (98.8%).<br />

• Patients treated with atovaquone/proguanil had a significantly shorter<br />

fever clearance time than patients treated with<br />

pyrimethamine/sulfadoxine (mean, 30.4 vs 44.9hrs; P


VII.<br />

Conclusions<br />

Antimalarial agents recommended in the suppression and treatment of malaria by the CDC include<br />

chloroquine products, hydroxychloroquine, mefloquine, primaquine, quinine, and the combination<br />

agent atovaquone-proguanil. Clinical comparative and efficacy studies show both combination<br />

agents are highly effective, while there are no significant differences between the agents in terms<br />

of fever and parasite clearance time, and cure rates.<br />

Therefore, all brand products within the class reviewed are comparable to each other and to the<br />

generics in the class and offer no significant advantage over other alternatives in general use.<br />

VIII. Recommendations<br />

No brand combination antimalarial agent is recommended for preferred status.<br />

300


References<br />

1 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

2 Shah, S., et al., Malaria Surveillance—United States, 2002. MMWR Surveill Summ, 2004. 53(1)<br />

p. 21-34.<br />

3 Centers for Disease Control and Prevention, Malaria Facts. Available at<br />

http://www.cdc.gov/malaria/facts.htm. Accessed September 2004.<br />

4 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

5 Centers for Disease Control and Prevention, Health Information for International Travel, 2003-<br />

2004. Available at http://www.cdc.gov/travel/diseases/malaria/index.htm. Accessed September<br />

2004.<br />

6 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

7 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

8 Centers for Disease Control and Prevention, Health Information for International Travel, 2003-<br />

2004. Available at http://www.cdc.gov/travel/diseases/malaria/index.htm. Accessed September<br />

2004.<br />

9 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

10 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

11 Gruber J. Hydroxychloroquine-Chloroquine; Pharmacokinetic Data. Available at<br />

http://www.lymenet.de/literatur/hydroxychloroquine.htm. Version: July 23, 2004. Accessed<br />

September 2004.<br />

12 Lariam ® [Medication Guide]. Nutley, NJ: F. Hoffmann-La Roche LTD; May 2004.<br />

13 The South African Medicines Formulary: Antiprotozoals, Part 2: Antimalarials. Available at<br />

http://web.uct.ac.za/depts/mmi/jmoodie/p01part2.html. Accessed September 2004.<br />

14 Daraprim® Tablet; Pharmacokinetic properties. Available at<br />

http://emc.medicines.or.uk/emc/assets/c/html/displaydoc.aspdocumentid=729. Accessed<br />

September 2004.<br />

15 Tatro, ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

16 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

17 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

18 Lariam ® [Medication Guide]. Nutley, NJ: F. Hoffmann-La Roche LTD; May 2004.<br />

19 Aralen ® [Prescribing Information]. New York, NY: Sanofi-Synthelabo; June 2003.<br />

20 Plaquenil ® [Prescribing Information]. New York, NY: Sanofi-Synthelabo; April 2002.<br />

21 Daraprim ® [Prescribing Information]. Research Triangle Park, NC: GlaxoSmithKline; March<br />

2003.<br />

22 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

23 Aralen ® [Prescribing Information]. New York, NY: Sanofi-Synthelabo; June 2003.<br />

24 Plaquenil ® [Prescribing Information]. New York, NY: Sanofi-Synthelabo; April 2002.<br />

25 Lariam ® [Medication Guide]. Nutley, NJ: F. Hoffmann-La Roche LTD; May 2004.<br />

26 Daraprim® Tablet; Posology and Method of Administration. Available at<br />

http://emc.medicines.or.uk/emc/assets/c/html/displaydoc.aspdocumentid=729. Accessed<br />

September 2004.<br />

27 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

28 MICROMEDEX ® Healthcare Series, Thomson MICROMEDIX, Greenwood Village, Colorado;<br />

Edition Expires 12-2004.<br />

29 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

301


30 Klasco, RK ed. USP DI ® Drug Information for the Health Care Professional. Thomson<br />

MICROMEDIX, Greenwood Village, Colorado; 2004.<br />

31 Ibrahim MH, Elbashir MI, Naser A, Aelbasit IA, Kheir NM, Adam I. Low-dose Quinine is<br />

effective in the treatment of Chloroquine-resistant Plasmodium falciparum malaria in Easter<br />

Sudan. Ann Trop Med Parasitol. 2004 Jul; 98(5): 441-5.<br />

32 White NJ, Krishna S, Waller D, Craddock C, Kwiatkowski D, Brewster D. Open comparison of<br />

intramuscular chloroquine and quinine in children with sever chloroquine-sensitive falciparum<br />

malaria. Lancet. 1989 Dec 2;2(8675): 1313-6.<br />

33 Baird JK, Fryauff DJ, Basri H, Bangs MJ, Subianto B, Wiady I, Purnomo, Leksana B, Masbar<br />

S, Richie TL, et al. Primaquine for prophylaxis against malaria among non-immune transmigrants<br />

in Irian Jaya, Indonesia. Am J trop Med Hyg, 1995 Jun; 52(6): 479-84.<br />

34 Mengesha T, Makonnen E. Comparative efficacy and safety of chloroquine and alternative<br />

Antimalarial drugs: a meta-analysis from six African countries. East Afr Med J. 1999 Jun: 76(6):<br />

314-9.<br />

35 Randrianarivelojosia M, Ratsimbasoa A, Randrianasolol L, Randrianarijaona A, Jambou R. Invitro<br />

sensitivity of Plasmodium falciparum in chloroquine, halofantrine, mefloquine and quinine in<br />

Madagascar. East Afr Me J. 2002 May; 79(5): 237-41.<br />

36 Schwartz E, Regev-Yochay G. Primaquine as prophylaxis for malaria for non-immune travelers:<br />

A comparison with mefloquine and doxycycline. Clin Infect Dis. 1999 Dec; 29(6): 1502-6.<br />

37 Kofoed K, Petersen E. The efficacy of chemoprophylaxis against malaria with chloroquine plus<br />

proguanil, mefloquine, and atovaquone plus proguanil in travelers from Denmark. J Travel Med.<br />

2003 May-Jun; 10(3): 150-4.<br />

38 Warhurst DC, Steele JC, Adagu IS, Craig JC, Cullander C. Hydroxychloroquine is much less<br />

active than chloroquine against chloroquine-resistant Plasmodium falciparum, in agreement with it<br />

physicochemical properties. J Antimicrob Chemother. 2003 Aug; 52(2): 188-93.<br />

39 Weiss WR, Oloo AJ, Johnson A, Koech D, Hoffman SL. Daily primaquine is effective for<br />

prophylaxis against falciparum malaria in Kenya: comparison with mefloquine, doxycycline, and<br />

chloroquine plus proguanil. J Infect Dis. 1995 Jun; 171(6): 1569-75.<br />

40 Fansidar ® [Product Information]. Nutley, NJ: F. Hoffmann-La Roche LTD; August 2004<br />

41 Centers for Disease Control and Prevention, Health Information for International Travel, 2003-<br />

2004. Available at http://www.cdc.gov/travel/diseases/malaria/index.htm. Accessed September<br />

2004.<br />

42 Centers for Disease Control and Prevention, Treatment Guidelines: Treatment of Malaria<br />

(Guidelines for Clinicians). Available at http://www.cdc.gov/malaria/pdf/treatmenttable.pdf.<br />

Accessed September 2004.<br />

43 MICROMEDEX ® Healthcare Series, Thomson MICROMEDIX, Greenwood Village, Colorado;<br />

Edition Expires 12-2004.<br />

44 Fansidar ® [Product Information]. Nutley, NJ: F. Hoffmann-La Roche LTD; August 2004<br />

45 Malarone ® [Prescribing Information]. Research Triangle Park, NC: GlaxoSmithKline;<br />

September 2004<br />

46 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

47 Tatro, ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

48 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

49 MICROMEDEX ® Healthcare Series, Thomson MICROMEDIX, Greenwood Village, Colorado;<br />

Edition Expires 12-2004.<br />

50 Kastrup EK, ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

51 Fansidar ® [Product Information]. Nutley, NJ: F. Hoffmann-La Roche LTD; August 2004<br />

52 Malarone ® [Prescribing Information]. Research Triangle Park, NC: GlaxoSmithKline;<br />

September 2004<br />

53 Fansidar ® [Product Information]. Nutley, NJ: F. Hoffmann-La Roche LTD; August 2004<br />

54 Malarone ® [Prescribing Information]. Research Triangle Park, NC: GlaxoSmithKline;<br />

September 2004<br />

55 Fansidar ® [Product Information]. Nutley, NJ: F. Hoffmann-La Roche LTD; August 2004<br />

56 Malarone ® [Prescribing Information]. Research Triangle Park, NC: GlaxoSmithKline;<br />

September 2004<br />

302


57 Klasco, RK ed. USP DI ® Drug Information for the Health Care Professional. Thomson<br />

MICROMEDIX, Greenwood Village, Colorado; 2004.<br />

58 Llanos-Cuentas A, Campos P, Clendenes M, Canfield CJ, Hutchinson DB. Atovaquone and<br />

proguanil hydrochloride compared with chloroquine or pyrimethamine/sulfadoxine for treatment<br />

of acuter Plasmodium falciparum malaria in Peru. Braz J Infect Dis. 2001 Apr; 5(2): 67-72.<br />

59 Mulenga M, Sukwa TY, Canfield CJ, Hutchinson DB. Atovaquone and proguanil versus<br />

pyrimethamine/sulfadoxine for the treatment of acute falciparum malaria in Zambia. Clin Ther.<br />

1999 May; 21(5): 841-52.<br />

60 Bustos DG, Canfield CJ, Canete-Miguel E, Hutchinson DB. Atovaquone-proguanil compared<br />

with chloroquine and chloroquine-sulfadoxine-pyrimethamine for treatment of acute Plasmodium<br />

falciparum malaria in the Philippines. J Infect Dis. 1999 Jun; 179(6): 1587-90.<br />

303


I. Overview<br />

Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

<strong>Pharmacotherapy</strong> <strong>Review</strong> of the Misc. Antiprotozoals<br />

AHFS 083092<br />

October 27, 2004<br />

Protozoal infections of the gastrointestinal tract occur worldwide and cause significant morbidity<br />

and mortality. Prevalence is higher in developing countries and areas unable to afford proper<br />

sanitation. In the United States, protozoal and other parasitic infections are most common in<br />

immigrants and recent travelers, institutional settings, daycare centers, pregnant women, and in<br />

immunocompromised patients. The drugs to be reviewed in this class are indicated for various<br />

parasitic and protozoal infections. Two of the drugs to be reviewed are also used to treat<br />

infections caused by anaerobic, gram negative, and gram positive bacteria. Pertinent parasites and<br />

protozoa will be discussed.<br />

Cryptosporidiosis<br />

Cryptosporidiosis is caused by the coccidian parasite Cryptosporidium parvum. The parasite is<br />

transmitted by ingestion of oocysts excreted in the feces of infected humans or animals. 1<br />

Transmission generally occurs through person-to-person contact, ingestion of contaminated<br />

drinking or recreational water, ingestion of contaminated food, from animal to person, or by direct<br />

contact with contaminated feces or environmental surfaces. 1 Cryptosporidium oocysts are highly<br />

resistant to chemical disinfectants used to treat drinking water and small numbers are even able to<br />

escape filtration. 1 Highest rates of occurrence are in persons aged 1-5 years and in patients with<br />

AIDS. 2<br />

In immunocompetent patients, Cryptosporidiosis is manifested as acute, self-limiting diarrhea<br />

lasting 7 to 14 days and may be accompanied by nausea, abdominal cramping, and low-grade<br />

fever. In the United States, approximately 2.5% of non-immunocompromised patients have<br />

oocyts of C. parvum and approximately 30% of non-immunocompromised patients are seropositive<br />

for C. parvum. Cryptosporidiosis was rarely reported until the beginning of the AIDS<br />

epidemic. In patients with AIDS, the infection is usually chronic and more severe. In the past,<br />

10-15% of patients with AIDS developed cryptosporidiosis. 2 However, with the introduction of<br />

highly active anti-retroviral therapy, the incidence has dropped.<br />

Giardiasis<br />

Giardiasis is caused by the protozoa Giardia lamblia (also known as Giardia intestinalis) and is<br />

the most common intestinal parasite in the United States. 3 Prevalence in the United States is as<br />

high as 3-13% and carrier rates are as high as 30-60% 5 Giardiasis is not associated with<br />

significant mortality except in children and pregnant women who can become extremely<br />

dehydrated. Person-to-person transmission of the cyst form of the protozoa is the most common<br />

means of infection. However, because it can survive in cyst form outside of the human body,<br />

Giardia can be found in soil, food, recreational water, or contaminated feces. The trophozoite<br />

form of the protozoa causes intestinal symptoms including diarrhea, flatulence, greasy stools,<br />

stomach cramps, and nausea. Symptoms usually occur 1 to 2 weeks after infection. 4 Anorexia,<br />

malaise, and weight loss are also common. Chronic infection can lead to nutritional deficiencies<br />

due to malabsorption.<br />

Amebiasis<br />

Amebiasis is caused by the protozoa Entamoeba histolytica. Entamoeba is transmitted in cyst<br />

form through contaminated water, food, or feces. The trophozoite form of the protozoa is<br />

responsible for symptomatic infection. E. histolytica infections can remain inside the intestine or<br />

they can spread to include abscesses of the perineum, genitalia, pericardium, peritoneum, liver,<br />

lungs, and the brain in rare cases. Clinical manifestations of intestinal infection range from vague<br />

304


complaints of abdominal discomfort and malaise to severe abdominal cramps, flatulence, and<br />

bloody diarrhea with mucus. 3 Fever occurs only in 10-30% of patients. In the United States,<br />

prevalence of Entamoeba infection is approximately 4%, although only 10% of persons who are<br />

infected have symptomatic disease. Institutionalized individuals (particularly the mentally<br />

retarded), homosexual males, immunosuppressed patients, and recent travelers are at highest risk<br />

for infection.<br />

Balantidiasis<br />

Balantidiasis is caused by the protozoa Balantidium coli. Balantidiasis is not very common in the<br />

United States, with an estimated prevalence of 1%. 6 Sporadic infections have been reported in<br />

institutionalized settings. Internationally, balantidiasis is associated with those who frequently<br />

come in contact with swine and generally demonstrates mortality only in patients who are<br />

malnourished or immunocompromised. Balantidiasis is transferred from person-to-person or<br />

animal to person in the cyst form and can exist outside of the human body in contaminated water<br />

or feces. Most infections are asymptomatic, but acute illness can present as diarrhea, mucus and<br />

blood in the stools, nausea, and intestinal colic.<br />

Trichomoniasis<br />

Trichomoniasis is a sexually transmitted protozoal infection caused by Trichomonas vaginalis. In<br />

the United States, trichomoniasis affects 2 to 3 million women annually. 7 Prevalence in men<br />

ranges from 8-31%. 7 The organism can survive for short period of time on moist surfaces, such as<br />

bathing or toilet articles. Symptoms in women range from none in asymptomatic carriers to<br />

severe pelvic inflammatory disease. Common symptoms include yellow to grayish vaginal<br />

discharge, vaginal odor, vaginal itching, and dysuria. 3 Infected pregnant women are 30% more<br />

likely than uninfected women to have a low birth weight infant. 7 Symptoms in men range from<br />

none in asymptomatic carriers to urethral discharge, pruritus, dysuria, urethritis, and prostatitis. 3<br />

Pneumocystis Carinii Pneumonia<br />

Pneumocystis carinii (also known as Pneumocystis jiroveci) is a unicellular eukaryote that is<br />

sometimes regarded as a protozoan because of its susceptibility to anti-protozoal drugs.<br />

Pneumocystis carinii pneumonia (PCP) is a major source of morbidity and mortality in<br />

immunocompromised patients. 8 It is the most common opportunistic respiratory infection in HIV<br />

patients and is also a pathogen in patients with a history of malignancy, autoimmune diseases, and<br />

organ transplant. 8 PCP is transmitted through inhalation of air-borne cysts that infect the<br />

respiratory tract. Most people in the United States show seroconversion by age 3, but only<br />

immunocompromised patients develop clinical disease. 8 Early in the HIV epidemic, 75% of HIV<br />

patients developed PCP at some point. With the addition of prophylactic therapy, the range is now<br />

10-20% with a mortality rate of 10-20% . 8 Since the additional of prophylactic therapy, incidence<br />

of PCP in other immunocompromised patients has dropped to 0-10% with a mortality rate of<br />

40%. 8 Classic symptoms of PCP include non-productive cough, fever, dyspnea, chest pain,<br />

sputum production, and tachypnea.<br />

305


This review encompasses all dosage forms and strengths. Table 1 lists the drugs included in this<br />

review.<br />

Table 1. Misc. Antiprotozoals in this <strong>Review</strong><br />

Generic Name Formulation Example Brand Name (s)<br />

Nitazoxanide Powder for Oral Suspension, Oral Tablet Alinia<br />

Metronidazole Oral Tablet, Oral Extended Release Tablet, Oral<br />

Capsule, Lyophilized Powder for Injection,<br />

Injection<br />

*Flagyl<br />

*Flagyl ER<br />

Flagyl IV, Metro IV, Flagyl IV RTU<br />

Protostat (D/c’d)<br />

Atovaquone Oral Suspension Mepron<br />

Trimetrexate<br />

Lyophilized Powder for Injection<br />

Neutrexin<br />

glucuronate<br />

Pentamidine<br />

Isethionate<br />

Solution for Inhalation, Injection<br />

NebuPent<br />

*Pentam 300<br />

Furazolidone Oral Tablets, Oral Liquid **Furoxone<br />

*Generic Available.<br />

** Per manufacturer, Furoxone has been unavailable for 2 years with no release date.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Cryptosporidiosis<br />

The current drug of choice for cryptosporidiosis in non-HIV infected patients is nitazoxanide. 9<br />

However, nitazoxanide has not been shown to be superior to placebo in HIV-infected patients. 9<br />

Nitazoxanide is the first drug to be approved for the treatment of cryptosporidiosis and the first to<br />

become available as a liquid for the treatment of giardiasis. 10 Nitazoxanide is approved for the<br />

treatment of diarrhea caused by Cryptosporidium parvum or Giardia lamblia in pediatric patients<br />

aged 1 through 11 years. 11 Nitazoxanide’s exact mechanism of action us unknown, but it is<br />

believed to interfere with anaerobic energy metabolism. Nitazoxanide inhibits the growth of<br />

Cryptosporidium oocysts and Giardia trophozoites. Nitazoxanide is also active against Isospora<br />

belli, Entamoeba histolytica, as well as various helminths. 10<br />

Prior to the introduction of nitazoxanide, paromomycin and azithromycin were used in the<br />

treatment of cryptosporidiosis, but without clearly demonstrated effectiveness. 10 In the treatment<br />

of giardiasis, metronidazole was the treatment of choice prior to the introduction of nitazoxanide.<br />

However, nitazoxanide has the advantage of being a liquid formulation, better tolerated, and has a<br />

shorter duration of treatment. 10<br />

In the guidelines for the diagnosis and management of foodborne illnesses, the CDC has<br />

recommendations for the diagnosis and treatment of illness caused by contaminated food sources.<br />

Specifically, paromomycin and nitazoxanide are recommended in severe illness of children aged<br />

1-11 years. 11 The CDC has also posted guidelines for the prevention of transmission through<br />

water and food sources, person-to-person transmission, animal-to-person transmission, and<br />

prevention of exposure in HIV infected persons. 1<br />

Giardiasis<br />

The drugs of choice for the treatment of giardiasis are currently metronidazole and nitazoxanide,<br />

with tinidazole and paromomycin as alternatives. 9 In the treatment of giardiasis, metronidazole<br />

was the treatment of choice prior to the introduction of nitazoxanide. However, nitazoxanide has<br />

the advantage of being a liquid formulation, better tolerated, and has a shorter duration of<br />

treatment. 10 In the guidelines for the diagnosis and management of foodborne illnesses, the CDC<br />

has recommendations for the diagnosis and treatment of illness caused by contaminated food<br />

sources. Metronidazole is listed as the treatment of choice for giardiasis. 12<br />

306


Amebiasis<br />

The drugs of choice for the treatment of amebiasis caused by Entamoeba histolytica vary<br />

depending on severity of disease. The recommended treatment for asymptomatic to mild intestinal<br />

disease is iodoquinol or paromomycin. 9 The drugs of choice for moderate to severe intestinal<br />

disease or for extraintestinal disease are metronidazole or tinidazole. 9 In the guidelines for the<br />

diagnosis and management of foodborne illnesses, the CDC has recommendations for the<br />

diagnosis and treatment of illness caused by contaminated food sources. Metronidazole and a<br />

luminal agent (iodoquinol or paromomycin) are listed as the treatments of choice for giardiasis. 12<br />

Balantidiasis<br />

The drug of choice for the treatment of balantidiasis is tetracycline with metronidazole and<br />

iodoquinol as alternatives. 9 Because tetracycline is not recommended in pregnant women or<br />

children due to potential discoloration of permanent teeth, metronidazole would be a useful<br />

alternative.<br />

Trichomoniasis<br />

The drugs of choice for the treatment of trichomoniasis are metronidazole or tinidazole. 9 Brigham<br />

and Woman’s Hospital has published guidelines for the diagnosis and treatment of common<br />

gynecological problems. Recommended treatment for the treatment of trichomoniasis is oral<br />

metronidazole. 13 During pregnancy, common practice is to consider deferring treatment until after<br />

the first trimester, although the CDC recommends pregnant women can be treated even in the first<br />

trimester to prevent adverse fetal outcome. 13<br />

Pneumocystis Carinii (Jiroveci) Pneumonia<br />

The drug of choice for the treatment of Pneumocystis carinii pneumonia (PCP) is<br />

trimethoprim/sulfamethoxazole (SMZ/TMP) with alternatives being primaquine plus clindamycin,<br />

trimethoprim plus dapsone, pentamidine, trimetrexate glucuronate, or atovaquone. The drug of<br />

choice for the primary and secondary prophylaxis of PCP is trimethoprim/sulfamethoxazole with<br />

alternatives being dapsone, dapsone plus pyrimethamine, pentamidine aerosol, or atovaquone. 9<br />

Guidelines exist for the prevention and treatment of PCP in persons infected with HIV, and in<br />

patients who have recently received solid organ, blood, or bone marrow transplants.<br />

The 2001 USPHS/DSA guidelines for the prevention of opportunistic infections in person infected<br />

with HIV, as well as additional sources, recommend PCP prophylaxis regimens in patients with<br />

one or more prior episodes of PCP, a CD4+ count of less than 200 cells/mm 3 , thrush associated<br />

14, 15<br />

with HIV, or unexplained fevers greater than 100 degrees F for more than two weeks. Of the<br />

drugs in this review, aerosolized pentamidine and atovaquone are alternative choices to the<br />

recommended regimen of SMZ/TMP. Primary prophylaxis can be discontinued in patients<br />

responding to highly active anti-retroviral therapies (HAART) who have had an increase in CD4+<br />

T-cell counts of greater than 200 cells/mm 3 for at least three months. 14 Of the drugs in this review,<br />

intravenous pentamidine, atovaquone, and trimetrexate glucuronate are alternative choices to the<br />

recommended regimen of SMZ/TMP.<br />

In the case of solid organ, blood, or bone marrow transplant recipients, the CDC recommends<br />

prophylaxis for PCP from the time of transplant to a minimum of six months for all patients, and<br />

greater than six months for those receiving immunosuppressive therapy or those with graft-versus<br />

host disease. 16,17 Of the drugs in this review, inhaled pentamidine is the best alternative to the<br />

preferred drug SMZ/TMP. 16<br />

307


III.<br />

Comparative Indications of the Misc. Anti-Protozoals<br />

Table 2 lists the FDA-approved indications for the miscellaneous anti-protozoal agents. Table 3<br />

further describes the major protozoal organisms and the treatments of choice.<br />

Table 2. FDA-Approved Indications for the Misc. Anti-Protozoals 11<br />

Drug Cryptosporidium<br />

parvum<br />

Giardia<br />

lamblia<br />

Entamoeba<br />

histolytica<br />

Trichomonas<br />

vaginalis<br />

Balantidium<br />

coli<br />

Nitazoxanide<br />

Metronidazole<br />

Atovaquone<br />

✔<br />

✔<br />

Misc. organisms (i.e.<br />

Bacteroides sp.,<br />

Fusobacterium sp.,<br />

Clostridium sp.,<br />

Eubacterium sp.<br />

Peptococcus sp.,<br />

Peptostreptococcus sp.)<br />

✔ ✔ ✔ ✔ ✔<br />

Pneumocystis<br />

carinii<br />

✔<br />

Vibrio<br />

cholerae<br />

Trimetrexate<br />

glucuronate<br />

✔<br />

Pentamidine<br />

Isethionate<br />

✔<br />

Furazolidone * ✔ ✔<br />

* Furazolidone is also active against various intestinal bacterial that cause diarrhea such as E. coli, Campylobacter, Enterobacter, Proteus,<br />

Salmonella, Shigella, and staphylococci.<br />

9, 11<br />

Table 3. Major Protozoal Infections, Causative Organisms, and Treatments of Choice<br />

Infection Organism Drug(s) of Choice (+ alternatives)<br />

Cryptosporidiosis<br />

Cryptosporidium<br />

parvum<br />

Nitazoxanide 1<br />

Giardiasis Giardia lamblia Metronidazole, Nitazoxanide (Tinidazole, Paromomycin,<br />

Furazolidone)<br />

Amebiasis<br />

Entamoeba<br />

histolytica<br />

Intestinal infection: Paromomycin, Iodoquinol<br />

Severe intestinal or extraintestinal infection:<br />

Metronidazole or Tinidazole followed by Paromomycin or<br />

iodoquinol)<br />

Balantidiasis Balantidium coli Tetracycline (Metronidazole, Iodoquinol)<br />

Trichomoniasis<br />

Pneumocystis carinii pneumonia<br />

(PCP)<br />

Trichomonas<br />

vaginalis<br />

Pneumocystis carinii<br />

Metronidazole, Tinidazole<br />

Treatment: SMZ/TMP (Primaquine plus Clindamycin,<br />

Trimethoprim plus Dapsone, intravenous Pentamidine,<br />

Atovaquone)<br />

Prophylaxis: SMZ/TMP (Dapsone, Dapsone plus<br />

Pyrimethamine, inhaled Pentamidine, Atovaquone)<br />

1<br />

Ntazoxanide is only indicated in infections in children age 1 through 11 years. Effectiveness in HIV-infected patients has not<br />

been consistently proven.<br />

308


IV.<br />

Pharmacokinetic Parameters of the Misc. Anti-Protozoals<br />

Nitazoxanide<br />

Metronidazole<br />

Atovaquone<br />

of Action<br />

Believed to be<br />

due to<br />

interference<br />

with the<br />

pyruvate:<br />

ferredoxin<br />

oxidoreductase<br />

enzymedependent<br />

electron<br />

transfer<br />

reaction,<br />

which is<br />

essential to<br />

anaerobic<br />

energy<br />

metabolism.<br />

Undergoes<br />

intracellular<br />

chemical<br />

reduction via<br />

mechanisms<br />

unique to<br />

anaerobic<br />

metabolism.<br />

Reduced<br />

metronidazole<br />

is cytotoxic<br />

and interacts<br />

with DNA to<br />

cause a loss of<br />

helical<br />

structure,<br />

strand<br />

breakage, and<br />

therefore<br />

inhibition of<br />

nucleic acid<br />

synthesis and<br />

cell death.<br />

Not fully<br />

known.<br />

Possibly cidal<br />

by inhibiting<br />

mitochondrial<br />

electrontransport<br />

chain<br />

at site of<br />

cytochrome<br />

bc1 complex,<br />

ultimately<br />

inhibiting the<br />

synthesis of<br />

nucleic acid<br />

and ATP.<br />

Well absorbed<br />

orally<br />

(at least 80%).<br />

Oral peak<br />

serum in 1 to<br />

2 hours.<br />

Plasma<br />

concentrations<br />

are<br />

proportional to<br />

dose.<br />

Oral<br />

suspension has<br />

two-fold<br />

bioavailability<br />

over oral<br />

tablets.<br />

Bioavailability<br />

of each<br />

increases twofold<br />

when<br />

given with<br />

meals (47%<br />

suspension<br />

and 23%<br />

tablet).<br />

Binding<br />

>99% Tizoxanide –<br />

eliminated in<br />

urine, bile<br />

and feces<br />

309<br />

Tizoxanide<br />

glucuronide<br />

– eliminated<br />

in urine and<br />

bile<br />

94% fecal.<br />

Active<br />

Metabolites<br />

Yes –<br />

tizoxanide<br />

and<br />

tizoxanide<br />

glucuronide<br />

Yes – 2-<br />

hydroymethyl<br />

Elimination<br />

Biliary;<br />


Trimetrexate<br />

glucuronate<br />

Pentamidine<br />

Isethionate<br />

(Intravenous)<br />

Pentamidine<br />

Isethionate<br />

(Inhaled)<br />

Furazolidone<br />

A folate<br />

antagonist that<br />

inhibits the<br />

enzyme<br />

dihydrofolate<br />

reductase,<br />

therefore<br />

disrupting<br />

DNA, RNA,<br />

and protein<br />

synthesis<br />

resulting in<br />

cell death.<br />

Not fully<br />

understood.<br />

May interfere<br />

with nuclear<br />

metabolism<br />

and inhibit the<br />

synthesis of<br />

DNA, RNA,<br />

proteins and<br />

phospholipids.<br />

Not fully<br />

understood.<br />

May interfere<br />

with nuclear<br />

metabolism<br />

and inhibit the<br />

synthesis of<br />

DNA, RNA,<br />

proteins and<br />

phospholipids<br />

Microbicidal.<br />

Minimizes<br />

development<br />

of resistant<br />

organisms by<br />

interfering<br />

with several<br />

bacterial<br />

enzyme<br />

systems. Also<br />

acts as an<br />

MAOI.<br />

Very<br />

lipophilic.<br />

Injectable. 10-<br />

30% excreted<br />

unchanged in<br />

urine, 50%<br />

excreted in<br />

feces as<br />

unchanged<br />

drug;


V. Drug Interactions of the Misc. Anti-Protozoals<br />

Administering atovaquone with food can increase its absorption two-fold.<br />

Metronidazole may interfere with chemical analyses for AST, ALT, LDH, triglycerides, and<br />

hexokinase glucose. Table 5 illustrates drug interactions with the agaents in this class.<br />

Table 5. Drug Interactions of the Misc. Anti-Protozoals 19<br />

Drug Significance Interaction Mechanism<br />

Nitazoxanide - Nitazoxanide and highly protein<br />

bound drugs<br />

Nitazoxanide is highly protein bound,<br />

therefore use with caution when administering<br />

with other highly protein-bound drugs due to<br />

Metronidazole<br />

Level 1 (delayed,<br />

major, established)<br />

Metronidazole and warfarin<br />

Metronidazole Level 2 (rapid,<br />

moderate,<br />

suspected)<br />

Metronidazole and ethanol<br />

Metronidazole Level 4 (delayed, Metronidazole and lithium<br />

moderate, possible)<br />

Metronidazole Level 4 (delayed, Metronidazole and hydantoins<br />

moderate, possible)<br />

Metronidazole Level 2 (delayed, Metronidazole and barbiturates<br />

moderate,<br />

suspected)<br />

Metronidazole Level 2 (delayed, Metronidazole and disulfiram<br />

moderate,<br />

suspected)<br />

Metronidazole Level 5 (delayed, Metronidazole and cimetidine<br />

minor, possible)<br />

Atovaquone - Atovaquone and highly protein<br />

bound drugs<br />

Atovaquone<br />

Level 2 (delayed,<br />

moderate,<br />

suspected)<br />

Atovaquone and zidovudine<br />

311<br />

competition for binding sites.<br />

Liver metabolism of warfarin may be<br />

decrease, therefore increasing the<br />

anticoagulant effect of warfarin.<br />

Disulfiram-like reaction due to accumulation<br />

of acetaldehyde by interference with the<br />

oxidation of alcohol.<br />

May cause serum lithium levels to increase.<br />

Phenytoin levels may increase due to<br />

decreased clearance.<br />

Metronidazole’s elimination can be<br />

accelerated, resulting in decreased plasma<br />

levels.<br />

May result in acute psychosis or confused<br />

state.<br />

Metronidazole serum levels may increase due<br />

to decreased clearance.<br />

Atovaquone is highly protein bound,<br />

therefore use with caution when<br />

administering with other highly proteinbound<br />

drugs due to competition for<br />

binding sites. Phenytoin has not been<br />

affected.<br />

Atovaquone can inhibit the glucuronidation of<br />

zidovudine, therefore elevating serum<br />

zidovudine concentrations.<br />

Atovaquone - Atovaquone and rifamycins Decrease in plasma concentrations of<br />

atovaquone and increase in plasma<br />

concentrations of rifampin.<br />

Atovaquone<br />

Trimetrexate<br />

glucuronate<br />

Trimetrexate<br />

glucuronate<br />

Pentamidine<br />

Isethionate<br />

(injectable)<br />

Furazolidone<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Atovaquone and etoposide<br />

- Trimetrexate and drugs that<br />

inhibit cytochrome P450 (i.e.<br />

erythromycin, fluconazole,<br />

rifampin, rifabutin)<br />

- Trimetrexate imidazole drugs<br />

(clotrimazole, ketoconazole,<br />

miconazole)<br />

Level 1 (delayed,<br />

major, suspected)<br />

Level 2 (rapid,<br />

moderate, probable)<br />

Pentamidine and ziprasidone<br />

Furazolidone and Ethanol<br />

Mechanism unknown. Atovaquone may<br />

elevate etoposide concentrations.<br />

Could result in trimetrexate toxicity, due to<br />

decreased metabolism.<br />

Could result in inhibited trimetrexate<br />

metabolism, resulting in toxicity.<br />

Prolongation of QT interval could result in<br />

cardiac arrythmias.<br />

Inhibition of enzymes responsible for<br />

conversion of acetaldehyde to acetate could


Furazolidone<br />

Furazolidone<br />

Furazolidone<br />

Furazolidone<br />

Furazolidone<br />

Level 1 (rapid,<br />

major, suspected)<br />

Level 2 (delayed,<br />

moderate,<br />

suspected)<br />

Level 4 (rapid,<br />

major, possible)<br />

Level 4 (rapid,<br />

major, possible)<br />

Level 4 (delayed,<br />

moderate, possible)<br />

Furazolidone and<br />

Sympathomimetics<br />

Furazolidone and Anorexiants<br />

Furazolidone and Levodopa<br />

Furazolidone and Meperidine<br />

Furazolidone and Tricyclic<br />

antidepressants<br />

cause a disulfiram-like reaction.<br />

Furazolidone may increase the pressor<br />

sensitivity to mixed and indirect-acting<br />

sympathomimetics possibly resulting in<br />

hypertension. Direct-acting sympathomimetics<br />

(eg, dobutamine [eg, Dobutrex]) are not<br />

affected.<br />

Increased sensitivity to pressor response of<br />

anorexiants due to MAO inhibition.<br />

Efficacy and adverse effects of levodopa could<br />

be increased due to inhibition of MAO.<br />

Unknown. Could include agitation, seizures,<br />

diaphoresis, fever, coma.<br />

Possible augmentation or release of amines in<br />

the CNS could cause variable effects including<br />

psychosis.<br />

VI.<br />

Adverse Drug Events of the Misc. Anti-Protozoal Agents<br />

Table 6. Common Adverse Events (%) Reported for the Misc. Anti-Protozoal Agents 11<br />

Adverse Event Nitazoxanide Metronidazole Atovaquone * Trimetrexate<br />

glucuronate **<br />

Body as a Whole<br />

Malaise<br />

Infection<br />

Influenza symptoms<br />

Moniliasis<br />

Myalgia<br />

Cardiovascular<br />

Edema<br />

Hypotension<br />

Hypertension<br />

Chest pain<br />

Digestive System<br />

Abdominal Pain<br />

Nausea / Vomiting<br />

Diarrhea<br />

Epigastric distress<br />

Appetite decrease<br />

Appetite increase<br />

Flatulence<br />

Metallic taste<br />

Dry mouth<br />

Central Nervous System<br />

Dizziness/Vertigo<br />

Fatigue<br />

Fever<br />

Headache<br />

Meningeal Signs<br />

Raised Intracranial<br />

Pressure<br />

Collapse<br />

Confusion<br />

Drowsiness<br />

Hepatic<br />

Abnormal LFTs (incr.)<br />

Hepatitis<br />

Jaundice<br />

Hepatic failure<br />

Skin and Appendages<br />

Alopecia<br />

Rash<br />

Sweat<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

7.8<br />

1.1<br />

2.1<br />

-<br />

-<br />

b<br />

b<br />

-<br />

-<br />

b<br />

-<br />

b<br />

1.1<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

7<br />

6<br />

3<br />

-<br />

-<br />

-<br />

-<br />

-<br />

4<br />

10<br />

4<br />

-<br />

-<br />

-<br />

-<br />

9<br />

2<br />

4<br />

-<br />

-<br />

18<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

312<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

Pentamidine<br />

isethionate<br />

(injectable)<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

0.9<br />

-<br />

-<br />

-<br />

5.9<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

1.7<br />

-<br />

8.7<br />

-<br />

-<br />

-<br />

-<br />

3.3<br />

-<br />

Pentamidine<br />

isethionate<br />

(inhaled)<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

b<br />

b<br />

b<br />

-<br />

b<br />

-<br />

-<br />

-<br />

b<br />

-<br />

b<br />

-<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

Furazolidone<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-


Pruritus b - - - - - -<br />

Hematologic<br />

Neutropenia<br />

Agranulocytosis<br />

Anemia<br />

Thrombocytopenia<br />

Leukopenia<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

b<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

1.2<br />

1.7<br />

2.8<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

Renal<br />

Abnormal kidney fxn<br />

Acute kidney failure<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

0.5<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

Metabolic<br />

Increased creatinine<br />

Electrolyte imbalance<br />

Hypoglycemia<br />

Hypocalcemia<br />

GU<br />

Vaginitis<br />

Genital pruritus<br />

Abnormal urine<br />

Dysmenorrhea<br />

UTI<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

15<br />

5<br />

3<br />

3<br />

2<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

23<br />

-<br />

2.4<br />

0.2<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

-<br />

-<br />

Respiratory<br />

Rhinitis<br />

URTI<br />

Sinusitis<br />

Pharyngitis<br />

Cough<br />

Other<br />

Angioedema<br />

Convulsions<br />

4<br />

4<br />

3<br />

3<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

bAdverse event reported; specific percentages not available<br />

• Percentages varied for atovaquone and trimetrexate adverse effects based upon each study and each drug being compared against. It is<br />

difficult to distinguish adverse events from underlying effects of HIV.<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

b<br />

b<br />

b<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

VII.<br />

Dosing and Administration for the Misc. Anti-Protozoals<br />

Agents9, 11<br />

Table 7. Dosing for the Anti-Protozoal<br />

Drug Availability Dose /Frequency/Duration<br />

Nitazoxanide 100mg/5ml powder Children 12 to 47 months: 5ml every 12 hours for 3 days<br />

for oral suspension,<br />

500mg tablet Children 4 to 11 years: 10ml every 12 hours for 3 days<br />

Metronidazole<br />

250mg oral tablet<br />

500mg oral tablet<br />

750mg extended<br />

release tablet<br />

375mg capsules<br />

500mg powder for<br />

injection<br />

5mg/ml injection<br />

Other Notes<br />

• Take with food.<br />

• Safety and efficacy has not been established for infants less than 1 year<br />

of age.<br />

Intestinal Amebiasis:<br />

750mg three times daily for 5 to 10 days<br />

Amebic liver abscess:<br />

500 or 750mg three times daily for 5 to 10 days<br />

Children: 35 to 50 mg/kg/24 hours (max 750mg/dose) in 3 divided doses for<br />

10 days<br />

Giardiasis:<br />

250mg three times daily for 5 days<br />

Children: 15mg/kg/day in three doses for 5 days<br />

Trichomoniasis:<br />

1 day treatment: 2gm either as single dose or in two divided doses in one day.<br />

7 day treatment: Adults: 250mg three times daily for 7 days<br />

313


Atovaquone<br />

Trimetrexate<br />

glucuronate<br />

Pentamidine<br />

Isethionate<br />

(Intravenous)<br />

Pentamidine<br />

Isethionate<br />

750mg/5ml<br />

suspension<br />

25mg powder for<br />

injection<br />

300mg injection<br />

300mg aerosol<br />

314<br />

Children: 5mg/kg/dose three times daily for 7 days<br />

Balantidiasis:<br />

As alternative to tetracycline: 750mg three times daily for 5 days<br />

Children: 35-50mg/kg/day in three doses for 5 days.<br />

Other Notes<br />

• Take with meals for snack to lessen GI irritation.<br />

• ER tablets should be taken on an empty stomach.<br />

• When used to treat trichomoniasis, treat sexual partner as well, even if<br />

asymptomatic.<br />

Avoid alcohol during treatment and for 4 days after.<br />

PCP prophylaxis:<br />

Age 13 to adult: 1500mg once daily with a meal.<br />

PCP treatment:<br />

Age 13 to adult: 750mg twice daily with food for 21 days<br />

Failing to take doses with food can result in a two-fold decrease in<br />

absorption.<br />

Trimetrexate must be given with concurrent leucovorin to avoid potentially<br />

serious or life-threatening toxicities. Leucovorin must be given daily during<br />

trimetrexate therapy and for 72 hours past the last dose.<br />

Moderate to severe PCP in adults >18 years old:<br />

45mg/m 2 once daily IV over 60 to 90 minutes. Leucovorin may be administered<br />

IV at a dose of 20mg/m 2 over 5 to 10 minutes every 6 hours for a total daily dose<br />

of 80mg/m 2 or orally as 4 doses of 20mg/m 2 spaced equally throughout the day.<br />

Round up dose to next 25mg increment. Length of therapy is 21 days of<br />

trimetrexate and 24 days of leucovorin.<br />

Toxicity grades 1 through four are listed below in ascending order:<br />

Neutrophils /mm 3 Platelets/mm 3 Trimetrexate Leucovorin<br />

>1,000 >75,000 45mg/m 2 once daily 20mg/m 2 q 6 h<br />

750-1000 50,000-75,000 45mg/m 2 once daily 40mg/m 2 q 6 h<br />

500-749 25,000-49,999 22mg/m 2 once daily 40mg/m 2 q 6 h<br />


(Inhaled)<br />

Furazolidone<br />

100mg tablets<br />

50mg/15ml liquid<br />

(Both unavailable<br />

with no release date)<br />

5-7 L/min.<br />

Average of 5mg/kg/day given in four divided doses. Do not exceed 8.8<br />

mg/kg/day due to nausea.<br />

Adults: 100mg four times daily<br />

Children: (5 years or older) 25 to 50 mg four times daily<br />

(1 to 4 years) 17 to 25 mg four times daily<br />

(1 month to 1 year) 8 to 17 mg four times daily.<br />

Avoid alcohol during treatment and for up to 4 days after.<br />

Avoid foods containing tyramine.<br />

Avoid OTC and prescription sympathomimetics.<br />

Special Dosing Considerations<br />

11, 18<br />

Table 8. Special Dosing Considerations for the Misc. Anti-Protozoal Agents<br />

Drug Renal Hepatic Pediatric Use Pregnancy<br />

Dosing Dosing<br />

Nitazoxanide No No Only indication is for children<br />

aged 1 through 11 years.<br />

Safety and effectiveness has<br />

not been established in<br />

patients < 1 year old.<br />

Metronidazole<br />

Reduce to<br />

50% of<br />

normal<br />

dose for<br />

GFR <<br />

10ml/min<br />

Yes –<br />

monitor<br />

for<br />

toxicity<br />

Atovaquone No Yes –<br />

use with<br />

caution<br />

Trimetrexate<br />

glucuronate<br />

Pentamidine<br />

Isethionate<br />

(Intravenous)<br />

Interrupt<br />

tx if<br />

serum<br />

creatinine<br />

>2.5mg/dl<br />

Interrupt<br />

tx if<br />

LFT’s <<br />

5 times<br />

upper<br />

limit of<br />

normal<br />

range.<br />

Safety and efficacy in<br />

children has not been<br />

established except in<br />

treatment of amebiasis.<br />

Newborns show a diminished<br />

capacity to eliminate.<br />

No information is available of<br />

the efficacy in pediatric<br />

patients. Clinical experience<br />

is limited to data in<br />

immunocompromised patients<br />

aged 1 month to 13 years.<br />

Safety and efficacy has not<br />

been established for patient <<br />

18 years of age. Data<br />

indicated two children treated<br />

successfully without adverse<br />

event.<br />

No No Data is limited, however<br />

dosing is the same as adults.<br />

Category<br />

B<br />

B<br />

C<br />

D<br />

C<br />

Can Drug Be Crushed/Stability<br />

Available in liquid. No data available<br />

on administration per tube.<br />

Tablets may be crushed and<br />

suspended in cherry syrup. Stable for<br />

30 days at ambient room temperature<br />

or refrigerated.<br />

IV form must be diluted or<br />

neutralized unless in RTU form.<br />

Reconstituted IV form is stable for 96<br />

hours when stored


than 1 month old due to<br />

possibility of hemolytic<br />

anemia.<br />

formulations.<br />

VIII. Comparative Effectiveness of the Misc. Anti-Protozoals<br />

Table 9. Additional Outcomes Evidence for the Misc. Anti-Protozoals<br />

Study Sample Treatment /<br />

Duration<br />

Ortiz JJ,<br />

et al. 20 n=110 Nitazoxanide for 3<br />

days vs.<br />

metronidazole for 5<br />

days for giardiasis<br />

Amadi B n=100 Nitazoxanide for<br />

et al. 21 cryptosporidiosis<br />

Colby C,<br />

et al. 22 n=39 Atovaquone vs.<br />

SMZ/TMP following<br />

autologous stem cell<br />

transplantation<br />

(ASCT)<br />

Dohn M, n=109 Atovaquone vs. IV<br />

et al. 23 pentamidine in the<br />

treatment of PCP<br />

El-Sadr , n=1057 Dapsone vs.<br />

et al. 24 atovaquone for PCP<br />

prophylaxis<br />

Results<br />

100 children with giardiasis were randomized to either a 3-day course of nitazoxanide<br />

or a 5-day course of metronidazole:<br />

• Diarrhea had resolved in 85 % of the nitazoxanide group vs. 80% for<br />

metronidazole.<br />

• Only mild, transient adverse events were reported.<br />

• Results: a 3-day course of nitazoxanide is as efficacious as a 5-day course of<br />

metronidazole in the treatment of giardiasis in children.<br />

50 HIV positive children and 50 HIV-negative children with C. parvum were<br />

randomly assigned to either nitazoxanide or placebo:<br />

• In HIV-negative children, diarrhea resolved in 56% of drug group vs. 23% of<br />

placebo.<br />

• In HIV-negative children, C. Parvum was eradicated form stool in 52% of drug<br />

group vs. 14% placebo.<br />

• HIV positive patients did not benefit from nitazoxanide.<br />

39 post-ASCT patients were studied. 16 patients received atovaquone and 19 patients<br />

received SMZ/TMP for the prevention of PCP:<br />

• None of the patients treated with atovaquone were removed from the study due<br />

to treatment associated adverse events. 40% of the patients randomized to<br />

SMZ/TMP dropped out of the study due to drug intolerance.<br />

• None of the patients treated developed PCP.<br />

• Atovaquone is better tolerated than SMZ/TMP in the prophylaxis of PCP after<br />

ASCT.<br />

109 patients with HIV and mild to moderate PCP received either atovaquone (n=56)<br />

or IV pentamidine (n=53) for 21 days:<br />

• More patients were successfully treated with atovaquone (57%) than with IV<br />

pentamidine (40%), but more patients failed to respond to atovaquone (29%)<br />

than pentamidine(17%).<br />

• Discontinuation of treatment due to adverse events was more common with<br />

pentamidine (36%) than with atovaquone (4%).<br />

• Atovaquone and IV pentamidine have similar efficacy in the treatment of PCP,<br />

but atovaquone may have fewer adverse events.<br />

1057 HIV-positive patients (298 had a history of PCP) were randomized to receive<br />

either daily atovaquone (536) or daily dapsone (521):<br />

• 122 of 536 atovaquone patients developed PCP vs. 135 of 521 dapsone patients.<br />

• Among the 546 patients who were receiving dapsone at base line, the relative<br />

risk of discontinuation because of adverse events was 3.78 for atovaquone as<br />

compared with dapsone, among those not receiving dapsone at base line, it was<br />

0.42.<br />

• Among patients who cannot tolerate trimethoprim-sulfamethoxazole, atovaquone<br />

and dapsone are similarly effective for the prevention of P. carinii pneumonia.<br />

Results support the continuation of dapsone prophylaxis among patients who are<br />

already receiving it. However, among those not receiving dapsone, atovaquone<br />

is better tolerated and may be the preferred choice for prophylaxis against P.<br />

carinii pneumonia.<br />

316


Chan C, et<br />

al. 25 n=549 Atovaquone<br />

suspension vs.<br />

pentamidine aerosol<br />

for PCP prophylaxis<br />

549 HIV-positive patients who were intolerant to SMZ and/or TMP were randomized<br />

to receive either atovaquone suspension or aerosolized pentamidine:<br />

• The incidence of treatment-limiting adverse events was significantly higher with<br />

atovaquone than with pentamidine.<br />

• There was no significant difference in the time using therapy.<br />

• Incidences of PCP and death were higher in patients receiving 750mg<br />

atovaquone than 1500mg atovaquone.<br />

• Atovaquone 1500mg daily had similar efficacy to that of aerosolized<br />

pentamidine.<br />

Additional Evidence<br />

Dose Simplification: In most cases, the antiprotozoal agents in this class are given as a single<br />

dose or for a brief duration (acute use), depending on the infection being treated. When given for<br />

longer durations, such as PCP prophylaxis, dosing is once daily.<br />

Stable Therapy: There is little evidence of resistance to antiprotozoal therapy in the literature.<br />

The majority of time that standard therapy was discontinued for a particular indication, the cause<br />

is intolerability to the drug of choice. There are many reports in the literature of resistance to<br />

anti-protozoal drugs, particularly in the treatment of trichomoniasis and giardiasis. Resistance<br />

against metronidazole is frequently reported, and there is cross-resistance within the family of 5-<br />

nitroimidazole drugs. There is also evidence that resistance to the drugs used to treat PCP is also<br />

developing, including SMZ/TMP, atovaquone and pentamidine.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal clinical<br />

literature relevant to use of the antiprotozoals and their impact on physician visits.<br />

IX.<br />

Conclusions<br />

Nitazoxanide is the first drug to be approved for Cryptosporidiosis. Although it does not seem to<br />

be anymore efficacious than metronidazole in the treatment of giardiasis, it does have the<br />

advantage of being a liquid formulation. Therefore, nitazoxanide’s primary use is in the treatment<br />

of Cryptosporidiosis. Treatment of this infectious disease is not applicable to the general<br />

population.<br />

In the treatment of giardiasis, metronidazole in the drug of choice, therefore furazolidone does not<br />

offer any advantage over metronidazole due to its current unavailability, brand name status, and<br />

drug interaction profile.<br />

In the treatment of Pneumocystis carinii pneumonia, SMZ/TMP is the drug of choice. However,<br />

due to intolerability, injectable pentamidine, atovaquone and trimetrexate are alternatives to<br />

standard therapy. Injectable pentamidine is available generically. Atovaquone and trimetrexate<br />

are not available generically. Injectable pentamidine and atovaquone show similar efficacy in the<br />

treatment of PCP, although atovaquone may be better tolerated. Trimetrexate has a disadvantage<br />

compared to atovaquone due to the need to administer with leucovorin and its inability to be used<br />

in pregnancy or in children and would be considered a third-line agent.<br />

In the prophylaxis of Pneumocystis carinii pneumonia, SMZ/TMP is the drug of choice. However,<br />

due to intolerability, aerosolized pentamidine or atovaquone are alternatives to standard therapy.<br />

Neither drug is available generically. Aerosolized pentamidine and atovaquone show similar<br />

efficacy to each other. Aerosolized pentamidine has the advantage of being better tolerated, but<br />

unlike systemic agents, offers no protection against other types of infections. 15 Treatment and<br />

prophylaxis therapies for Pneumocystis carinii pneumonia are not applicable to the general<br />

population.<br />

The primary role of the drugs in this class, as pertinent to general use in the population, is for the<br />

treatment of giardiasis, amebiasis, balantidiasis, and trichomoniasis. The drug in this class<br />

indicated for these infectious diseases is metronidazole. Metronidazole is available as a generic<br />

317


formulation. Therefore, all brand anti-protozoal agents in this class are comparable to each other<br />

and to the generics and OTC products in the class and offer no significant clinical advantage over<br />

other alternatives in general use. Additionally, the therapies for Cryptosporidiosis and<br />

Pneumocystis carinii pneumonia are not within the scope of general use in the population, and<br />

should be available for their indicated special needs/circumstances via medical justification<br />

through the prior authorization process.<br />

X. Recommendations<br />

No brand antiprotozoal agent is recommended for preferred status.<br />

318


References<br />

1. Juranek DD. Centers for Disease Control and Prevention (CDC). Cryptosporidiosis: Sources of<br />

Infection and Guidelines for Prevention. Accessed at www.cdc.gov September 2004.<br />

2. Eisen, D. Cryptosporidiosis. Accessed at www.emedicine.com. Topic #484. Last updated<br />

March 2004.<br />

3. Anandan JV. Parasitic Diseases. In: <strong>Pharmacotherapy</strong>. A Pathophysiologic Approach.<br />

Dipiro JT, Talbert RL, Yee GC, et al. Eds. Elsevier Science. New York. 1989. Pg. 1197-1198.<br />

4. Centers for Disease Control and Prevention (CDC). Giardia Infection: Fact Sheet for the<br />

General Public. Accessed at www.cdc.gov September 2004.<br />

5. Pennardt, A. Giardiasis. Accessed at www.emedicine.com. Topic #215. Last updated June<br />

2004.<br />

6. Chijide, VM. Balantidiasis. Accessed at www.emedicine.com Topic # 203. Last updated March<br />

2002.<br />

7. Perkins AM. Trichomoniasis. Accessed at www.emedicine.com Topic # 2308. Last updated<br />

August 2004.<br />

8. Schreibman TS. Pneumocystis Carinii Pneumonia. Accessed at www.emedicine.com Topic #<br />

1850. Last updated August 2004.<br />

9. Abramowicz, M, Editor. The Medical Letter on Drugs and Therapeutics, Drugs for Parasitic<br />

Infections. The Medical Letter, Inc. New York August 2004.<br />

10. Abramowicz, M, Editor. The Medical Letter on Drugs and Therapeutics, Nitazoxanide (Alinia)<br />

– A New Anti-protozoal Agent. The Medical Letter, Inc. New York April 2003.<br />

11. Kastrup EK, Ed. Drug Facts and Comparisons. Facts and Comparisons. St. Louis. 2004.<br />

12. Centers for Disease Control and Prevention (CDC). Guidelines for the Diagnosis and<br />

Management of Foodborne illnesses. MMWR Apr 16 2004;53;RR-4.<br />

13. Lifford, K, et al. Brigham and Women’s Hospital: Common gynecological problems: a guide to<br />

diagnosis and treatment. Boston (MA) Brigham and Woman’s Hospital; 2002.<br />

14. Centers for Disease Control and Prevention (CDC). Guidelines for preventing opportunistic<br />

infections in HIV infected persons. MMWR Jun 2002 14;51 (RR8)1-52.<br />

15. Chaisson RE,Bishai W. The Management of Pneumocystis carinii, Toxoplasmosis, and HSV<br />

infections in patients with HIV disease. Last updated July 1999. Accessed at www.medscape<br />

.com September 2004.<br />

16. Centers for Disease Control and Prevention (CDC). Guidelines for preventing opportunistic<br />

infections among hematopoietic stem cell transplant recipients. MMWR Oct 20 2000;49;RR-<br />

10.<br />

17. Cincinnati Children’s Hospital Medical Center. Evidence based clinical practice guidelines for<br />

pneumocystis carinii pneumonia prophylaxis following solid organ or blood and marrow<br />

transplants. Cincinnati, OH. 2001 Jan 12.<br />

18. Micromedex, Healthcare Series;DrugDex Drug Evaluations 2004.<br />

19. Tatro, Ed. Drug Interaction Facts. Facts and Comparisons. St. Louis. 2004.<br />

20. Ortiz JJ, Ayoub A, Gargala G, et al. Randomized clinical study of nitazoxanide compared to<br />

metronidazole in treatment of symptomatic giardiasis in children from northern Peru. Aliment<br />

Pharmacol Ther Sept 2001;15(9):1409-1415.<br />

21. Amadi B, Mwiya M, Musuku J, et al. Effect of nitazoxanide on morbidity and mortality in<br />

Zambian children with cryptosporidiosis. Lancet Nov 2, 2002;360(9343):1375-1380.<br />

22. Colby C, McAfee S, Sackstein R, et al. A prospective randomized trial comparing toxicity and<br />

safety of atovaquone with SMZ/TMP as PCP prophylaxis following ASCT. Bone Marrow<br />

Transplant Oct 1999;24(8):897-902.<br />

23. Dohn MN, Weinberg WG, Torres RA, et al. Oral atovaquone compared with intravenous<br />

pentamidine for PCP pneumonia in patients with AIDS. Ann Intern Med 1994 Aug;121(3):174-<br />

180.<br />

24. El-Sadr WM, Murphy RL, Yurik TM, et al. Atovaquone compared with dapsone for the<br />

prevention of PCP in patients with HIV who cannot tolerate sulfonamides or trimethoprim. N<br />

Eng J Med Dec 1998;339(26):1889-95.<br />

25. Chan C, Montaner J, Lefebvre E, et al. Atovaquone suspension compared with aerosolized<br />

pentamidine for prevention of PCP in HIV-infected subjects intolerant of SMZ and/or TMP. J<br />

of Infec Dis 1999;180:369-376.<br />

319


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

New Drug <strong>Pharmacotherapy</strong> <strong>Review</strong><br />

Climara Pro (Estradiol/Levonorgestrel Transdermal System)<br />

Estrogen Replacement Products, Combination Agents, AHFS 681604<br />

October 27, 2004<br />

I. Overview<br />

Multiple hormone replacement treatments are available for women, including combination agents for<br />

use in women with a uterus, to avoid risk of endometrial cancer. Progestins are combined with<br />

estrogens for three reasons: 1) to decrease the risk of estrogen-induced irregular bleeding, endometrial<br />

hyperplasia and carcinoma; 2) to protect against breast cancer; and 3) to enhance estrogen prophylaxis<br />

of osteoporosis. 1<br />

The estradiol / levonorgestrel (Climara Pro) transdermal system is the second approved and available<br />

combination estrogen/progestin transdermal system. The new transdermal system is available as<br />

0.045mg/day estradiol and 0.015mg/day of levonorgestrel. This review encompasses all dosage forms<br />

and strengths of the new drug.<br />

The estradiol / norethindrone acetate (Combipatch) system was included in the original estrogen<br />

combination agent class review in March 2004. The original review of the estrogen replacement<br />

products, combination agents, is available in full for reference in Appendix 1. Other oral<br />

estrogen combination agents are available with estradiol, conjugated estrogen, and esterified<br />

estrogens. There are generic alternatives on the market for one oral estrogen combination<br />

product (Estratest / H.S.); however, no generic alternatives are available for the transdermal<br />

estrogen replacement combination products. This review encompasses all dosage forms and<br />

strengths.<br />

II.<br />

Current Treatment Guidelines<br />

The reported endometrial cancer risk among unopposed estrogen users is about 2 to 12 fold greater<br />

than in non-users, and appears dependent on duration of treatment and on estrogen dose. In<br />

addition, the greatest risk is associated with prolonged use, with increased risks of 15 to 24 fold<br />

for five to ten years or more. 2<br />

Information from the Women’s Health Initiative (WHI) trial about estrogens and progestins<br />

continues to have significant public health implications for postmenopausal women. The<br />

combined estrogen/progestin arm of the WHI study was stopped early because the increased risk<br />

of breast cancer and cardiovascular events exceeded the specified benefits. Additional results<br />

from the study are presented in Table 1. Many women’s health organizations now suggest that<br />

ERT / HRT be used only for management of vasomotor symptoms, using the lowest dose for the<br />

shortest duration. Topical vaginal products should especially be considered when ERT / HRT is<br />

only being considered for the treatment of vaginal atrophy. In addition, labeling for some oral<br />

combination estrogens has been updated to reflect the following changes: 2<br />

♦<br />

Estrogens and progestins should not be used for the prevention of cardiovascular<br />

disease.<br />

Since the March P&T meeting, results of the estrogen-alone arm of the Women’s Health Initiative<br />

(WHI) have suggested similar results as found earlier with the combination estrogen agents;<br />

estrogen-alone hormone therapy had no effect on coronary heart disease risk but increased the risk<br />

of stroke in postmenopausal women. The WHI also found that estrogen-alone therapy<br />

significantly increased the risk of deep vein thrombosis, had no significant effect on the risk of<br />

breast or colorectal cancer, and reduced the risk of hip and other fractures. 3<br />

320


As a result of requests for information on the approved labeling for hormone replacement<br />

products, and information from the WHI studies, the Food and Drug Administration (FDA) has<br />

established a website on estrogen and progestin therapies for postmenopausal women. 4 The<br />

website at www.fda.gov contains information for patients and healthcare professionals including<br />

product labeling, FDA Talk Papers, treatment guidelines, and additional resources for relating<br />

women’s health information.<br />

Table 1. Relative and Absolute Risk Seen in the CE/MPA Substudy of WHI A 5, 6<br />

Event<br />

Relative Risk CE/MPA vs.<br />

Placebo at 5.2 Years (95%<br />

Placebo<br />

n=8,102<br />

CE/MPA<br />

n=8,506<br />

CI*)<br />

Absolute Risk per 10,000 Person-Years<br />

CHD Events<br />

Non-fatal MI<br />

CHD death<br />

1.29 (1.02-1.63)<br />

1.32 (1.02-1.72)<br />

1.18 (0.70-1.97)<br />

30<br />

23<br />

6<br />

37<br />

30<br />

7<br />

Invasive Breast Cancer B 1.26 (1.00-1.59) 30 38<br />

Stroke 1.41 (1.07-1.85) 21 29<br />

Pulmonary Embolism 2.13 (1.39-3.25) 8 16<br />

Colorectal Cancer 0.63 (0.43-0.92) 16 10<br />

Endometrial Cancer 0.83 (0.47-1.47) 6 5<br />

Hip Fracture 0.66 (0.45-0.98) 15 10<br />

Death Due to Other Cause 0.92 (0.74-1.14) 40 37<br />

Global Index C 1.15 (1.03-1.28) 151 170<br />

Deep Vein Thrombosis D 2.07 (1.49-2.87) 13 26<br />

Vertebral Fractures D 0.66 (0.44-0.98) 15 9<br />

Other Osteoporotic Fractures D 0.77 (0.69-0.86) 170 131<br />

CE 0.625mg Conjugated Estrogens<br />

MPA 2.5mg Medroxyprogesterone<br />

A Adapted from JAMA 2002;288:321-333.<br />

B Includes metastatic and non-metastatic breast cancer with the exception of in situ breast cancer.<br />

C A subset of the events was combined in a “global index”, defined as the earliest occurrence of CHD events, invasive breast cancer,<br />

stroke, pulmonary embolism, endometrial cancer, colorectal cancer, hip fracture, or death due to other causes.<br />

D Not included in the Global index.<br />

* Nominal confidence intervals unadjusted for multiple looks and multiple comparisons.<br />

III.<br />

Indications<br />

The estradiol / levonorgestrel (Climara Pro) transdermal system is indicated in women with an<br />

intact uterus, for the treatment of moderate to severe symptoms associated with menopause. 6<br />

In comparison, estradiol / norethindrone (Combipatch) has indications for the treatment of<br />

vasomotor symptoms, atrophic vaginitis, hypogonadism, primary ovarian failure, and castration.<br />

Most of the oral estrogen combination agents are indicated for the treatment of vasomotor<br />

symptoms, atrophic vaginitis, and osteoporosis prevention.<br />

321


IV.<br />

Pharmacokinetics<br />

5, 6, 8<br />

Table 2. Pharmacokinetic Parameters of Select Combination Estrogen Agents<br />

Drug<br />

Time to Reach Peak Protein Metabolism Elimination Half-Life (T 1/2 )<br />

Estradiol /<br />

norethindrone<br />

(Activella)<br />

Estradiol /<br />

levonorgestrel<br />

(Climara Pro)<br />

Estradiol /<br />

norethindrone<br />

(Combipatch )<br />

Esterified<br />

estrogens /<br />

methyltestosterone<br />

(Estratest)<br />

Ethinyl estradiol /<br />

norethindrone<br />

(FemHRT)<br />

Concentration (T max )<br />

Estradiol- 5-8 hours<br />

Norethindrone- 0.5-<br />

1.5 hours<br />

Estradiol-36 hours<br />

Levonorgestrel-48<br />

hours<br />

Estradiol- 12-24<br />

hours<br />

Norethindrone- 24<br />

hours<br />

Esterified estrogens-<br />

NA<br />

Methyltestosterone-<br />

NA<br />

V. Drug Interactions<br />

Binding<br />

98% First-pass in<br />

the liver<br />

Primarily<br />

urine<br />

Estradiol- 12-14 hours<br />

Norethindrone- 8-11 hours<br />

- Liver Urine Estradiol- 3 ± 0.67 hours<br />

Levonorgestrel- 28 ± 6.4<br />

hours<br />

- Skin, Liver Urine Estradiol- 2-3 hours<br />

Norethindrone- 6-8 hours<br />

98% First-pass in<br />

the liver<br />

1-2 hours >95% First-pass in<br />

the liver<br />

Primarily<br />

urine<br />

Primarily<br />

Urine<br />

Esterified estrogens-NA<br />

Testosterone-10-<br />

100minutes<br />

Ethinyl Estradiol- 23.9<br />

hours<br />

Norethindrone- 13.3 hours<br />

In-vivo and in-vitro studies have shown that estrogens are metabolized partially by cytochrome<br />

P450 3A4. 6, 7 This includes the Climara Pro transdermal system. Therefore, inducers or inhibitors<br />

of CYP3A4 may affect estrogen drug metabolism. Inducers of CYP3A4 such as St. John’s Wort,<br />

phenobarbital, carbamazepine, and rifampin may reduce plasma concentrations of estrogens,<br />

possibly resulting in a decrease in therapeutic effects and/or changes in the uterine bleeding<br />

profile. CYP3A4 inhibitors such as erythromycin, clarithromycin, ketoconazole, itraconazole,<br />

ritonavir, and grapefruit juice may increase plasma concentrations of estrogens and may result in<br />

side effects. Additionally, levonorgestrel is metabolized through hydroxylation by the following<br />

cytochrome P450 enzymes: 3A, 2E, and 2C. Inducers or inhibitors of these enzymes may either,<br />

respectively, decrease the therapeutic effects or result in side effects. Dosage adjustments<br />

(increased or decreased) may be necessary to manage the induction or inhibition of estrogens<br />

when given in combination with interacting drugs.<br />

As all estrogens are metabolized similarly, drug interactions are common with these agents as a<br />

class. Most importantly, there are no major severity (level 1) drug interactions that have been<br />

documented with estrogens.<br />

VI.<br />

Adverse Drug Events<br />

As a result of the WHI results and current treatment guidelines, women should be treated with the<br />

lowest estrogen dose possible, to minimize risk of adverse events. Table 3 lists the reported<br />

adverse events in >3% of patients.<br />

Additionally, the irritation potential of the estradiol / levonorgestrel transdermal system was<br />

assessed in a 3-week irritation study. 6 Visual assessments of irritation were made on day 7 of each<br />

wear period, and were compared for a Climara Pro placebo patch and a Climara placebo patch.<br />

Mean irritation scores were 0.13 (week 1), 0.12 (week 2), and 0.06 (week 3) for the Climara Pro<br />

placebo and 0.20 (week 1), 0.26 (week 2), and 0.12 (week 3) for the Climara placebo. No<br />

irritation scores were greater than 2 at time in any subject during the study. In controlled trials,<br />

withdrawals due to application site reactions occurred in 2.1% of patients in the symptom study<br />

and in 8.5% of patients in the 1-year endometrial protection study. In comparison, use of<br />

322


Combipatch in studies did not result in any discontinuations due to application site reactions, of<br />

which occurred only in 4% of patients. 8<br />

Table 3. Clinical Adverse Events with Estradiol / Levonorgestrel Transdermal 6<br />

Adverse Event<br />

Climara Pro<br />

n=212<br />

Estradiol (E 2)<br />

n=204<br />

Body as a Whole<br />

Abdominal Pain<br />

Accidental Injury<br />

Back Pain<br />

Flu Syndrome<br />

Infection<br />

Pain<br />

9 (4.2)<br />

7 (3.3)<br />

13 (6.1)<br />

10 (4.7)<br />

7 (3.3)<br />

11 (5.2)<br />

11 (5.4)<br />

6 (2.9)<br />

12 (5.9)<br />

13 (6.4)<br />

10 (4.9)<br />

13 (6.4)<br />

Cardiovascular<br />

Hypertension 7 (3.3) 9 (4.4)<br />

Digestive<br />

Flatulence 8 (3.8) 5 (2.5)<br />

Metabolic and Nutritional<br />

Edema<br />

Weight Gain<br />

8 (3.8)<br />

6 (2.8)<br />

5 (2.5)<br />

10 (4.9)<br />

Musculoskeletal<br />

Arthralgia 9 (4.2) 10 (4.9)<br />

Nervous System<br />

Depression<br />

Headache<br />

Respiratory<br />

Bronchitis<br />

Sinusitis<br />

Upper Respiratory Infection<br />

Skin and Appendages<br />

Application Site Reaction<br />

Breast Pain<br />

Rash<br />

Urogenital<br />

Urinary Tract infection<br />

Vaginal Bleeding<br />

Vaginitis<br />

12 (5.7)<br />

11 (5.2)<br />

9 (4.2)<br />

8 (3.8)<br />

28 (13.2)<br />

86 (40.6)<br />

40 (18.9)<br />

5 (2.4)<br />

7 (3.3)<br />

78 (36.8)<br />

4 (1.9)<br />

7 (3.4)<br />

14 (6.9)<br />

7 (3.4)<br />

12 (5.9)<br />

26 (12.7)<br />

69 (33.8)<br />

20 (9.8)<br />

10 (4.9)<br />

8 (3.9)<br />

44 (21.6)<br />

6 (2.9)<br />

VII.<br />

Dosing and Administration<br />

Use of estrogen alone or in combination with a progestin should be limited to the lowest effective<br />

dose available and for the shortest duration consistent with treatment goals and risks for the<br />

individual woman. All patients should be reevaluated on a 3-6 month basis to determine if<br />

treatment is still necessary. Table 4 compares the dosing regimens for both available estrogen<br />

combination transdermal systems.<br />

Table 4. Dosing for the Transdermal Estrogen Combination Systems 6, 8<br />

Drug Dose/Frequency/Duration Available strengths<br />

Estradiol/Levonorgestrel 1 patch once weekly for 0.045mg estradiol /0.015mg<br />

(Climara Pro)<br />

Estradiol/Norethindrone<br />

(Combipatch)<br />

28 days<br />

1 patch twice a week for<br />

28 days<br />

levonorgestrel per day patch<br />

0.05mg estradiol/ 0.14mg<br />

norethindrone per day patch<br />

0.05mg<br />

estradiol/0.25mg<br />

norethindrone per day patch<br />

323


VIII. Effectiveness<br />

Multiple studies have evaluated the efficacy of transdermal estrogen combination systems. These<br />

studies are presented in Table 6. Additionally, the incidence of endometrial hyperplasia during<br />

continuous combined treatment with the estradiol / levonorgestrel (Climara Pro) transdermal<br />

system was evaluated in a study of 412 postmenopausal women. Results are illustrated in Table 5.<br />

Table 5. Incidence of Endometrial Hyperplasia During Continuous Combined Treatment<br />

with the Estradiol / Levonorgestrel Transdermal System 6<br />

0.045mg Estradiol /<br />

0.015mg Levonorgestrel<br />

(Climara Pro)<br />

n=210<br />

Estradiol (E 2 ) 0.045mg<br />

n=202<br />

# of patients with biopsies at ≥<br />

124 139<br />

6 months 1<br />

# of patients with biopsies at 1<br />

102 110<br />

year 2<br />

# (%) of patients with<br />

0 (0%) 19 (17.3%)<br />

hyperplasia 3<br />

95% Confidence interval 0-3.55% 9.75-24.79%<br />

n=number of intent-to-treat subjects.<br />

1<br />

Defined as at least 180 days of treatment.<br />

2<br />

Defined as at least 323 days of treatment.<br />

3<br />

Includes hyperplasia occurring at any time after initiation of treatment as a proportion of patients with biopsies at 1<br />

year.<br />

Table 6. Efficacy of the Transdermal Estrogen Combination Systems<br />

Study Sample Duration Results<br />

Estradiol / levonorgestrel<br />

transdermal patch vs. placebo for<br />

vasomotor symptoms 6 n=183 12 week<br />

randomized,<br />

placebo<br />

In evaluating the efficacy of the 0.045mg estradiol / 0.030mg<br />

levonorgestrel transdermal system when administered once<br />

weekly for moderate to severe vasomotor symptoms:<br />

controlled<br />

trial<br />

• The estradiol / levonorgestrel transdermal system<br />

was shown to be statistically better than placebo at<br />

weeks 4 and 12 for relief of both the number and<br />

Oral conjugated estrogen plus<br />

medroxyprogesterone vs.<br />

transdermal estradiol plus oral<br />

medroxyprogesterone on<br />

hypertriglyceridemia 9 n=61 3month,<br />

randomized<br />

trial<br />

Transdermal estradiol /<br />

levonorgestrel on bleeding pattern<br />

in postmenopausal women 10 n=468 4 week,<br />

randomized,<br />

multicenter<br />

study<br />

severity of moderate to severe hot flashes.<br />

Women participants included those who had received oral<br />

conjugated estrogen (0.625mg) plus 2.5mg<br />

medroxyprogesterone daily for 12 months, and developed<br />

triglyceride concentrations exceeding 150mg/dl. Patients<br />

were then randomly assigned to continue their current<br />

regimen or change to transdermal estradiol plus the same dose<br />

of medroxyprogesterone. Results indicated:<br />

• Serum concentrations of triglyceride and very-lowdensity<br />

lipoprotein triglyceride decreased<br />

significantly after changing to transdermal estradiol<br />

(triglyceride, from 226.0 +/- 43.9 to 110.5 +/- 44.1<br />

mg/dL, P < 0.01).<br />

• No changes were seen in concentrations of lowdensity<br />

lipoprotein cholesterol or high-density<br />

lipoprotein cholesterol.<br />

In evaluating the bleeding pattern and acceptability with use<br />

of transdermal estradiol alone for 2 weeks, followed by<br />

transdermal estradiol / levonorgestrel for 2 weeks:<br />

• The occurrence of cyclic bleeds was dose-dependent,<br />

with an increase at higher dosages; the frequency of<br />

a cyclic bleed per treatment cycle was 40% in the<br />

50/10 micrograms/24 h patch, 62% in the 75/15<br />

324


Transdermal estradiol /<br />

n=293<br />

levonorgestrel in 3 strengths (E 2<br />

0.045mg/day with 0.015, 0.030,<br />

or 0.040mg/day) 11<br />

Two<br />

prospective<br />

multicenter,<br />

double-blind,<br />

randomized,<br />

controlled<br />

trials<br />

micrograms/24 h patch and 76% in the 100/20<br />

micrograms/24 h patch.<br />

• The incidence of intermittent bleeding also increased<br />

with higher doses, from 22% in the 50/10 group to<br />

35% in the 100/20 group; 20% of women in the<br />

50/10 group did not bleed at all; the corresponding<br />

figures for the 75/15 and 100/20 groups were 7%<br />

and 0%, respectively.<br />

• Time of onset of cyclic bleeding was constant in all<br />

groups. The mean duration of cyclic bleeding was<br />

constant within each group, but increased from 4.4<br />

days in the 50/10 to 6.3 days in the 100/20 group.<br />

• The regularity and predictability of cyclic bleeding<br />

were high in all groups. Recurrence of cyclic bleeds<br />

was acceptable for most women (90%).<br />

Study 1: Patients received transdermal estradiol 0.045mg/day<br />

with 0.030 or 0.040mg/day levonorgestrel, or placebo for 3<br />

28-day cycles.<br />

Results:<br />

• Transdermal estradiol 0.045mg/day with 0.030 and<br />

0.040mg/day of levonorgestrel significantly<br />

decreased the number and severity of hot flushes<br />

when compared with placebo.<br />

• Symptom relief was seen as early as 2 weeks post<br />

treatment.<br />

n=845<br />

Efficacy and tolerability of<br />

transdermal estradiol /<br />

levonorgestrel as HRT in<br />

postmenopausal women 12 - 12 week<br />

double-blind,<br />

placebo<br />

controlled<br />

treatment<br />

phase,<br />

followed by a<br />

12 week<br />

open, follow-<br />

Study 2: Patients received transdermal estradiol<br />

0.045mgmg/day with 0.015, 0.030, or 0.040mg/day<br />

levonorgestrel, or transdermal estradiol 0.045mg/day as<br />

monotherapy for thirteen 28-day treatment cycles.<br />

Results:<br />

• No women receiving transdermal estradiol /<br />

levonorgestrel developed endometrial hyperplasia<br />

compared with 19 (12.8%) who received<br />

transdermal estradiol 0.045 mg/day alone (p <<br />

0.001 for each dose).<br />

• Significant improvements from baseline in scores<br />

on the Women's Health Questionnaire for<br />

vasomotor symptoms, sleep problems, sexual<br />

function, cognitive difficulties, and total score were<br />

noted at all or most time points with both<br />

transdermal estradiol / levonorgestrel and with<br />

estradiol alone.<br />

• Application-site reactions, vaginal hemorrhage,<br />

and breast pain were the most common adverse<br />

events reported with transdermal E2/LNG. The<br />

proportion of women with amenorrhea increased<br />

over time in all treatment groups in study 2.<br />

In evaluating the efficacy and tolerability of a 7-day<br />

transdermal sequential estradiol / levonorgestrel patch (2<br />

weeks of estradiol monotherapy patch followed by 2 weeks of<br />

the combination patch) with that of placebo in women aged<br />

40-65 with an intact uterus:<br />

• The sequential use of a 7-day estradiol patch and a 7-<br />

day estradiol / levonorgestrel patch was superior to<br />

placebo in reducing menopausal symptoms, and was<br />

well tolerated.<br />

325


up phase • At the end of the treatment phase, there was a<br />

statistically significant reduction in the Kupperman<br />

Index score (a measure of menopause symptoms)<br />

versus placebo (P


References<br />

1. Mullins PM, Pugh MC, Moore AO. Hormone Replacement Therapy. In: <strong>Pharmacotherapy</strong>. A<br />

Pathophysiologic Approach. Dipiro JT, Talbert RL, Yee GC, et al. Eds. Appleton & Lange.<br />

Connecticut. 1997. Pg. 1635-46.<br />

2. Prempro and Premphase ® [package insert]. Philadelphia, PA: Wyeth Pharmaceuticals Inc.; 2003.<br />

3. P&T Community. WHI study finds no heart disease benefit, increased stroke risk with estrogen<br />

alone. Available at: www.ptcommunity.com. Accessed June 2, 2004.<br />

4. Estrogen and estrogen with progestin therapies for postmenopausal women. U.S. Food and Drug<br />

Administration. Available at: www.fda.gov/cder/drug/infopage/estrogens_progestins. Accessed<br />

June 2, 2004.<br />

5. Kastrup EK, Ed. Drug Facts and Comparisons. Facts & Comparisons. St. Louis. 2004.<br />

6. Climara Pro [package insert]. Montville, NJ: Berlex;2003.<br />

7. Tatro DS, Ed. Drug Interaction Facts. Facts & Comparisons. St. Louis. 2004.<br />

8. Murray L, Senior Editor. Package Insert for Combipatch. In: Physicians’ Desk Reference, PDR<br />

Edition 58, 2004. Thomson PDR. Montvale, NJ. 2004.<br />

9. Sanada M, Tsuda M, Kodama I, et al. Substitution of transdermal estradiol during oral estrogenprogestin<br />

therapy in postmenopausal women: effects on hypertriglyceridemia. Menopause<br />

2004;11(3):331-336.<br />

10. Van de Weijer PH, Sturdee DW, von Holst T. Estradiol and levonorgestrel: effects on bleeding<br />

pattern when administered in a sequential combined regimen with a new transdermal patch.<br />

Climacteric 2002 Mar;5(1):36-44.<br />

11. Shulman LP, Yankov V, Uhl K. Safety and efficacy of a continuous once-a week 17beta-estradiol<br />

/ levonorgestrel transdermal system and its effects on vasomotor symptoms and endometrial safety<br />

in postmenopausal women: the results of two multicenter, double-blind, randomized, controlled<br />

trials. Menopause 2002 May-Jun;9(3):195-207.<br />

12. von Holst T, Salbach B. Efficacy of a new 7-day transdermal sequential estradiol / levonorgestrel<br />

patch in women. Maturitas 2002 Mar 25;41(3):231-42.<br />

13. Siseles NO, Benencia H, Mesch V, et al. Once and twice a week transdermal estradiol delivery<br />

systems: clinical efficacy and plasma estrogen levels. Climacteric 1998 Sep;1(3):196-201.<br />

327


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

New Drug <strong>Pharmacotherapy</strong> <strong>Review</strong>-<br />

Symbyax (Olanzapine and Fluoxetine capsules)<br />

AHFS Class 281604<br />

October 27, 2004<br />

I. Overview<br />

The depressive phase of bipolar disorder is associated with substantial morbidity, functional<br />

impairment, suicide, and other causes of excess mortality. 1 Patients with bipolar disorder<br />

experience depressive symptoms more than three times longer than they experience manic<br />

symptoms. 2 Treatment options for bipolar depression remain somewhat limited. Lithium or<br />

lamotrigine are currently considered first-line pharmacologic options for bipolar depression, based<br />

on the 2002 American Psychiatric Association (APA) treatment guidelines. (See “Evidence Based<br />

Medicine and Current Treatment Guidelines” below.) These agents are effective, but are not<br />

always associated with complete response, and in some cases are associated with treatmentlimiting<br />

side effects. 2 Overall, the treatment of bipolar depression is understudied, both in absolute<br />

terms and in comparison with the treatment of acute bipolar mania. 1 Since publication of the 2002<br />

APA guidelines, several new clinical studies regarding the acute treatment and prevention of<br />

bipolar depression have been published that may impact future treatment recommendations. 1<br />

Symbyax ® is a combination of two psychotropic agents, olanzapine and fluoxetine, indicated for<br />

the treatment of depressive episodes associated with bipolar disorder. Fluoxetine is a selective<br />

serotonin reuptake inhibitor (SSRI), while olanzapine has high affinity binding to serotonin,<br />

dopamine, muscarinic, histamine, and alpha 1 -adrenergic receptors. Although the exact mechanism<br />

of this combination product is unknown, it is proposed that the activation of three monoaminergic<br />

neural systems (serotonin, norepinephrine, and dopamine) is responsible for its enhanced<br />

antidepressant effect. 3<br />

Efficacy of the combination of olanzapine and fluoxetine in the treatment of bipolar I depression<br />

has been demonstrated in two clinical trials of identical study design (data were pooled and<br />

published in one article). 2 In these studies, olanzapine monotherapy significantly improved<br />

symptoms of bipolar depression, and the olanzapine/fluoxetine combination had an even stronger<br />

antidepressant effect without a greater risk of switch into mania.<br />

Olanzapine/fluoxetine combination is currently available as the brand product Symbyax ® and is<br />

available as 6mg/25mg, 6mg/50mg, 12mg/25mg, and 12mg/50mg capsules (mg olanzapine/mg<br />

fluoxetine). AHFS class 281604 was originally reviewed in July 2003, and this review has been<br />

included in the Appendix for reference. This review encompasses all dosage forms and strengths<br />

of the new drug.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

According to the APA 2002 treatment guidelines, the first-line pharmacological treatment for<br />

bipolar depression is the initiation of either lithium or lamotragine. 4 As an alternative, especially<br />

for more severely ill patients, some clinicians will initiate simultaneous treatment with lithium and<br />

an antidepressant. When an acute depressive episode does not respond to initial treatment at<br />

optimal doses, next steps include adding lamotrigine (if not used initially), bupropion, or<br />

paroxetine. Alternative next steps include adding a different SSRI, venlafaxine, or a monoamine<br />

oxidase inhibitor (MAOI). Antidepressant monotherapy is not recommended.<br />

The use of antidepressants in bipolar disorder is controversial. The APA guidelines recommend a<br />

cautious and limited, rather than routine, use of antidepressants in combination with mood<br />

stabilizers due to a risk of switching into mania and long-term worsening of bipolar illness. 5 On<br />

328


the other hand, the results of recent studies lend insight into both the degree to which mood<br />

stabilizers reduce the risk of switching associated with antidepressant treatment, and the<br />

magnitude of risk of depressive relapse after antidepressant discontinuation. 1<br />

III.<br />

Indications<br />

Olanzapine/fluoxetine combination is indicated for the treatment of depressive episodes associated<br />

with bipolar disorder. Its effectiveness for maintaining antidepressant response in this patient<br />

population beyond 8 weeks has not been established in controlled clinical studies.<br />

IV.<br />

Pharmacokinetics<br />

Concomitant administration of fluoxetine and olanzapine has resulted in small increases in the<br />

mean maximum concentration of olanzapine (≈16%), increases in the mean area under the curve<br />

(≈17%), and a small decrease in mean apparent clearance of olanzapine (≈16%). 3 The terminal<br />

half-life is not affected, so the time to reach steady state should not be altered. The overall steadystate<br />

plasma concentrations of olanzapine and fluoxetine when given as the combination in<br />

therapeutic dose ranges were comparable with those typically attained with each of the<br />

monotherapies. The observed small change in olanzapine clearance likely reflects the inhibition of<br />

a minor metabolic pathway for olanzapine via CYP2D6 by fluoxetine, a potent CYP2D6 inhibitor,<br />

and was not deemed clinically significant. Therefore, the pharmacokinetics of the individual<br />

components is expected to reasonably characterize the overall pharmacokinetics of the<br />

combination. 3<br />

Table 1. Pharmacokinetic Parameters of Olanzapine/Fluoxetine 3<br />

Olanzapine<br />

Fluoxetine<br />

Tmax 4 hr 6 hr<br />

Food Effect<br />

Does not affect rate or extent of • Delays absorption by 1-2 hrs<br />

absorption<br />

• No affect on systemic bioavailability<br />

Protein Binding 93% 94.5%<br />

Metabolism<br />

Highly metabolized by:<br />

Extensive, via CYP2D6, to norfluoxetine<br />

• Direct glucuronidation<br />

(active) and other metabolites<br />

• Cytochrome P450 1A2 and 2D6<br />

(minor), and flavin-containing<br />

monooxygenase system<br />

Elimination • Elimination half-life ≈ 21 to 54<br />

hrs<br />

• Metabolites excreted in urine<br />

(57%) and feces (30%)<br />

Elimination half-life<br />

• Parent drug: 1 to 3 days (acute) and 4 to 6<br />

days (chronic)<br />

• Active metabolite: 4 to 16 days<br />

Metabolites excreted in urine (57%) and feces<br />

(30%)<br />

329


V. Drug Interactions<br />

Because the metabolism of fluoxetine involves the CYP2D6 system, concomitant therapy with<br />

drugs also metabolized by this enzyme system may lead to drug interactions.<br />

Table 2. Clinically Significant Drug Interactions 3,6<br />

Drug Interaction Management<br />

Alprazolam<br />

Fluoxetine inhibits alprazolam<br />

metabolism<br />

Monitor for signs of alprazolam<br />

intoxication. Reduce alprazolam dose;<br />

switch to a different benzodiazepine<br />

with less interaction potential (e.g.,<br />

Antihypertensive agents<br />

Carbamazepine<br />

Clozapine<br />

Diazepam<br />

ECT<br />

Ethanol<br />

Haloperidol<br />

Levodopa, dopamine<br />

agonists<br />

Lithium<br />

MAOI<br />

NSAID/aspirin<br />

Phenytoin<br />

Olanzapine may induce<br />

hypotension, thereby enhancing the<br />

effects of antihypertensive agents<br />

Carbamazepine significantly<br />

increases olanzapine clearance and<br />

may reduce efficacy.<br />

Fluoxetine decreases<br />

carbamazepine metabolism, thus<br />

increasing carbamazepine levels.<br />

Fluoxetine decreases clozapine<br />

metabolism, thus increasing<br />

clozapine levels.<br />

Co-administration with olanzapine<br />

may potentiate olanzapine-induced<br />

orthostatic hypotension; Fluoxetine<br />

may prolong diazepam half-life<br />

Rare reports of prolonged seizures<br />

in patients on fluoxetine receiving<br />

ECT<br />

Co-administration may potentiate<br />

sedation and orthostatic<br />

hypotension.<br />

Elevated haloperidol blood levels<br />

with fluoxetine<br />

Olanzapine may antagonize the<br />

effects of these agents<br />

Both increased and decreased<br />

lithium levels have been observed<br />

with concomitant fluoxetine<br />

therapy. Lithium toxicity and<br />

serotonergic effects have been<br />

reported.<br />

Serious, sometimes fatal, reactions<br />

have been reported in patients<br />

receiving combination therapy and<br />

in patients receiving an MAOI<br />

after recently discontinuing<br />

fluoxetine.<br />

Increased risk of bleeding during<br />

concurrent use with SSRIs<br />

Elevated plasma levels of<br />

phenytoin with clinical toxicity due<br />

to inhibition of phenytoin<br />

metabolism by fluoxetine<br />

330<br />

lorazepam)<br />

Consider lower initial doses.<br />

Advise patients of risk, especially during<br />

initial dose titration<br />

Monitor for olanzapine efficacy and<br />

adjust dose as needed. Monitor<br />

carbamazepine levels and for signs of<br />

carbamazepine toxicity. Adjust<br />

carbamazepine doses accordingly.<br />

Monitor for signs of clozapine toxicity.<br />

Reduce clozapine dose if indicated<br />

Consider lower initial doses.<br />

Advise patients of risk, especially during<br />

initial dose titration<br />

Use cautiously in patients with history<br />

of seizures or with conditions that<br />

potentially lower seizure threshold.<br />

Avoid combination if at all possible.<br />

Due to risk of haloperidol toxicity and<br />

QT interval prolongation, concurrent<br />

administration is not recommended.<br />

Monitor patients for efficacy of antiparkinsonim<br />

agents.<br />

Monitor lithium levels.<br />

Use is contraindicated with, and for 5<br />

weeks after discontinuation of<br />

fluoxetine.<br />

Monitor for signs of bleeding.<br />

Monitor phenytoin levels with initiation<br />

of fluoxetine and periodically to assure<br />

stability. Also monitor levels for several<br />

weeks after discontinuation of


Drug Interaction Management<br />

fluoxetine. Adjust phenytoin dose<br />

accordingly.<br />

Pimozide<br />

Possible additive effects with Avoid concurrent use.<br />

fluoxetine on the QT interval<br />

leading to bradycardia<br />

Sumatriptan<br />

Rare postmarketing reports of<br />

weakness, hyperreflexia, and<br />

incoordination with fluoxetine<br />

Closely monitor patient for adverse<br />

effects.<br />

Thioridazine<br />

Tricyclic<br />

antidepressants (TCA)<br />

Tryptophan<br />

Warfarin<br />

Narrow therapeutic<br />

index drugs<br />

metabolized by<br />

CYP2D6 (e.g.,<br />

flecainide, vinblastine)<br />

Omeprazole, rifampin,<br />

other CYP1A2 or<br />

glucoronyl transferase<br />

inducers<br />

Digitoxin, other drugs<br />

tightly bound to plasma<br />

proteins<br />

Elevated plasma levels of<br />

thioridazine with resultant increase<br />

in potential of dose-related QT c<br />

interval prolongation, which is<br />

associated with serious ventricular<br />

arrhythmias and sudden death<br />

Increased TCA plasma levels >2-<br />

to 10-fold due to inhibition of<br />

metabolism by fluoxetine<br />

Adverse reactions including<br />

agitation, restlessness, and<br />

gastrointestinal distress, possibly<br />

due to additive effects with<br />

fluoxetine<br />

Altered anticoagulant effects<br />

including increased bleeding due to<br />

inhibition of warfarin metabolism<br />

by fluoxetine<br />

Increased plasma concentration of<br />

object drug due to inhibition of<br />

metabolism by fluoxetine<br />

Increased olanzapine clearance<br />

resulting in decreased plasma<br />

levels<br />

Shift in plasma concentrations of<br />

object drug potentially resulting in<br />

an adverse effect<br />

Use is contraindicated with, and for 5<br />

weeks after discontinuation of<br />

fluoxetine.<br />

Monitor TCA plasma level and reduce<br />

dose if indicated.<br />

Monitor patients for signs of serotonin<br />

syndrome.<br />

Closely monitor INR and patient for<br />

signs of increased anticoagulation.<br />

Consider dose reduction of the object<br />

drug, or initiate at low end of dose range<br />

if patient already receiving fluoxetine, or<br />

has received in the previous 5 weeks.<br />

Monitor for olanzapine efficacy and<br />

adjust dose as needed.<br />

Monitor for increased digoxin levels and<br />

for signs and symptoms of toxicity.<br />

CYP = cytochrome P450; ECT = electroconvulsive therapy; MAOI = monoamine oxidase inhibitor; NSAID = non-steroidal antiinflammatory<br />

drug; TCA = tricyclic antidepressant<br />

IV.<br />

Adverse Drug Events<br />

In short-term, controlled premarketing studies, 10% of patients receiving olanzapine/fluoxetine<br />

discontinued treatment due to adverse events compared with 4.6% for placebo. The most<br />

commonly observed adverse events associated with olanzapine/fluoxetine (≥5% and at least twice<br />

that for placebo) were: asthenia, edema, increased appetite, peripheral edema, pharyngitis,<br />

somnolence, thinking abnormal, tremor, and weight gain. 3 Table 3 lists adverse events reported in<br />

≥2% of patients receiving olanzapine/fluoxetine in controlled trials (with an incidence of twice or<br />

more that for placebo).<br />

331


Table 3. Treatment-Emergent Adverse Events: Incidence in Controlled Clinical Trials 3<br />

Adverse Event (%) Symbyax ®<br />

Bipolar Depression<br />

(n = 86)<br />

Symbyax ®<br />

Controlled<br />

(n = 571)<br />

Placebo<br />

(n = 477)<br />

Body as a Whole<br />

Asthenia 13 15 3<br />

Accidental injury 5 3 2<br />

Fever 4 3 1<br />

Cardiovascular System<br />

Hypertension 2 2 1<br />

Tachycardia 2 2 0<br />

Digestive System<br />

Diarrhea 19 8 7<br />

Dry mouth 16 11 6<br />

Increased appetite 13 16 4<br />

Tooth disorder 1 2 1<br />

Metabolic and Nutritional Disorders<br />

Weight gain 17 21 3<br />

Peripheral edema 4 8 1<br />

Edema 0 5 0<br />

Musculoskeletal System<br />

Joint disorder 1 2 1<br />

Twitching 6 2 1<br />

Arthralgia 5 3 1<br />

Nervous System<br />

Somnolence 21 22 11<br />

Tremor 9 8 3<br />

Thinking abnormal 6 6 3<br />

Lidibo decreased 4 2 1<br />

Hyperkinesia 2 1 1<br />

Personality disorder 2 1 1<br />

Sleep disorder 2 1 1<br />

Amnesia 1 3 0<br />

Respiratory System<br />

Pharyngitis 4 6 3<br />

Dyspnea 1 2 1<br />

Special Senses<br />

Ambylopia 5 4 2<br />

Ear pain 2 1 1<br />

Otitis media 2 0 0<br />

Speech disorder 0 2 0<br />

Urogenital System<br />

Abnormal ejaculation 7 2 1<br />

Impotence 4 2 1<br />

Anorgasmia 3 1 0<br />

The “Bipolar Depression” column shows the incidence of adverse events with Symbyax ® in the bipolar depression studies. The<br />

“Symbyax ® -Controlled” column shows the incidence in the controlled Symbyax ® studies. The placebo column shows the incidence in<br />

the pooled controlled studies that included a placebo arm.<br />

332


Selected Warnings and Precautions 3<br />

• Clinical worsening and suicide risk: Patients treated with antidepressants should be observed<br />

closely for clinical worsening and suicidality, especially at the beginning of therapy or at the<br />

time of dose changes. (See “FDA Public Health Advisory March 22, 2004” below)<br />

• Hyperglycemia and Diabetes Mellitus: Hyperglycemia, in some cases extreme and associated<br />

with ketoacidosis or hyperosmolar coma or death, has been reported in patients treated with<br />

atypical antipsychotics, including olanzapine, either alone or concomitantly with fluoxetine.<br />

(See “Consensus Statement on Antipsychotic Drugs and Obesity and Diabetes”)<br />

• Orthostatic Hypotension: Orthostatic systolic blood pressure decrease of ≥30 mm Hg occurred<br />

in 7.3%, 1.4%, and 1.4% of the olanzapine/fluoxetine, olanzapine, and placebo groups,<br />

respectively in bipolar depression studies. Use with caution in patients with known<br />

cardiovascular disease, cerebrovascular disease, or conditions that predispose patients to<br />

hypotension.<br />

• Mania/Hypomania: There was no statistically significant difference in the incidence of manic<br />

events between olanzapine/fluoxetine- and placebo-treated patients. However, the limited<br />

controlled trial experience of olanzapine/fluoxetine in bipolar depression and the cyclical<br />

nature of bipolar disorder warrant close monitoring for the development of symptoms of<br />

mania/hypomania during treatment with olanzapine/fluoxetine.<br />

• Weight gain: The mean weight increase for olanzapine/fluoxetine-treated patients was<br />

statistically significantly greater than placebo-treated (3.6 kg vs. –0.3 kg) and fluoxetinetreated<br />

(3.6 kg vs. –0.7 kg) patients, but not significantly different from olanzapine-treated<br />

patients (3.6 kg vs. 3.0 kg). Fourteen percent of olanzapine/fluoxetine-treated patients met the<br />

criterion for having gained >10% of their baseline weight, compared with


Table 4. Monitoring protocol for patients on atypical antipsychotics 9 *<br />

Baseline 4 weeks 8 weeks 12 weeks Quarterly Annually Every 5<br />

years<br />

Personal/family history X X<br />

Weight (BMI) X X X X X<br />

Waist circumference X X<br />

Blood pressure X X X<br />

Fasting plasma glucose X X X<br />

Fasting lipid profile X X X<br />

*More frequent assessments may be warranted based on clinical status<br />

Subsequent to the publication of the consensus statement, the Food and Drug Administration<br />

(FDA) asked all manufacturers of atypical antipsychotic medications to add a warning statement<br />

to their product labeling describing the increased risk of hyperglycemia and diabetes in patients<br />

taking these medications. 10<br />

VII.<br />

Dosing and Administration<br />

Olanzapine/fluoxetine should be administered once daily in the evening, generally beginning with<br />

the 6mg/25mg capsule. Dosage adjustments, if indicated, can be made according to efficacy and<br />

tolerability. Antidepressant efficacy was demonstrated in a dose range of olanzapine 6 to 12mg<br />

and fluoxetine 25 to 50mg. The safety of doses above 18mg/75mg has not been evaluated in<br />

clinical trials. 3<br />

Special Dosing Considerations: 3<br />

• The starting dose of olanzapine/fluoxetine 6mg/25mg should be used for patients with a<br />

predisposition to hypotensive reactions, patients with hepatic impairment, or patients who<br />

exhibit a combination of factors that may slow the metabolism of the drug (female gender,<br />

geriatric age, nonsmoking status). Dose escalation, when indicated, should be performed with<br />

caution in these patient populations.<br />

• Neonates exposed to fluoxetine or other SSRIs late in the third trimester have developed<br />

complications requiring prolonged hospitalization, respiratory support, and tube feeding. The<br />

physician may consider tapering fluoxetine in the third trimester.<br />

• Symptoms associated with discontinuation of fluoxetine and other SSRIs have been reported<br />

(dysphoric mood, irritability, agitation, dizziness, sensory disturbances, anxiety, confusion,<br />

headache, lethargy, emotional lability, insomnia, and hypomania). Patients should be<br />

monitored for these symptoms. A gradual reduction in the dose rather than abrupt cessation is<br />

recommended whenever possible.<br />

• The long elimination half-lives of fluoxetine and norfluoxetine assure that active drug will<br />

persist in the body for weeks, even when dosing is stopped. This is of potential consequence<br />

when drug discontinuation is required or when drugs are prescribed that might interact with<br />

fluoxetine and norfluoxetine following the discontinuation of fluoxetine.<br />

VIII. Effectiveness<br />

The efficacy of olanzapine and olanzapine-fluoxetine combination in the treatment of bipolar I<br />

depression was evaluated in an 8-week, multicenter, randomized, double-blind, placebo-controlled<br />

study. 2 A total of 833 patients were assigned to receive placebo (n=377), olanzapine (n=370) or<br />

olanzapine-fluoxetine combination (n=86; 6/25, 6/50, or 12/50mg/d administered as separate<br />

capsules taken together once daily in the evening). Eligible subjects were adults (age ≥18 years)<br />

who met DSM-IV criteria for bipolar I disorder, were depressed, had a Montgomery-Ashberg<br />

Depression Rating Scale (MADRS) total score of at least 20, and a history of at least one previous<br />

manic or mixed episode of sufficient severity to require treatment with a mood stabilizer or an<br />

antipsychotic agent. The primary measure of efficacy was the change in MADRS total score from<br />

baseline to week 8. Secondary efficacy measures included the Clinical Global Impressions<br />

Bipolar Version-Severity of Depression scale (CGI-BP-S), the Young Mania Rating Scale<br />

(YMRS), and the Hamilton Anxiety Rating scale (HAM-A) scores. Rates of and times to response<br />

334


(≥50% improvement in MADRS total score from baseline to an end point and completion of at<br />

least 4 weeks of study) and remission (MADRS score of 12 or less at an endpoint and completion<br />

of at least 4 weeks of study) were also assessed.<br />

Patients taking olanzapine-fluoxetine had the highest rate of completion (64.0%) compared with<br />

olanzapine (48.4%) or placebo (38.5%). From week 1 to the end of the study, the olanzapine and<br />

combination groups demonstrated significantly greater improvements in MADRS total scores than<br />

those receiving placebo (p


IX.<br />

Conclusions<br />

The combination of olanzapine and fluoxetine has demonstrated efficacy in the treatment of<br />

bipolar I depression in two clinical trials of identical study design (data were pooled and published<br />

in one article). 2 In these studies, olanzapine therapy significantly improved symptoms of bipolar<br />

depression, and the olanzapine/fluoxetine combination had an even stronger antidepressant effect<br />

(but not statistically significant) without a greater risk of switch into mania.<br />

The response rates achieved with the olanzapine/fluoxetine combination in these clinical trials<br />

appear similar to those achieved with lamotrigine, and possibly superior to those achieved with<br />

divalproex sodium in respective, placebo-controlled trials of those agents. 2,13,14 Such comparisons<br />

are limited, however, by methodological and sample differences between trials.<br />

The adverse event profile for the olanzapine/fluoxetine combination in clinical trials was similar to<br />

that of olanzapine monotherapy but also included higher rates of nausea and diarrhea. In addition,<br />

both active treatment groups were associated with significant weight gain, potentially clinically<br />

significant orthostatic hypotension, and elevations in nonfasting blood glucose and cholesterol<br />

levels.<br />

Use of the olanzapine/fluoxetine combination beyond 8 weeks has not been studied, nor has any<br />

advantage of the combination product over co-administration of the two ingredients separately (as<br />

in the clinical trials) been demonstrated. Furthermore, the question remains as to whether similar<br />

results could be achieved using other atypical antipsychotic agents in combination with a selective<br />

serotonin reuptake inhibitor or other antidepressants.<br />

Therefore, olanzapine/fluoxetine combination (Symbyax®) is comparable to the other brands in<br />

this class and to the generics and OTC products in this class and offers no significant advantage<br />

over other alternatives in general use.<br />

X. Recommendations<br />

No brand of olanzapine/fluoxetine is recommended for preferred status.<br />

336


References<br />

1. Keck PE, Nelson EB, McElroy SL. Advances in the pharmacologic treatment of bipolar<br />

depression. Biol Psychiatry. 2003;53:671-679.<br />

2. Tohen M, Vieta E, Calabrese J, et al. Efficacy of olanzapine and olanzapine-fluoxetine<br />

combination in the treatment of bipolar I depression. Arch Gen Psychiatry 2003;60(11):1079-<br />

88.<br />

3. Eli Lilly and Company. Symbyax (olanzapine and fluoxetine HCl) prescribing<br />

information. Indianapolis (IN): March 2004.<br />

4. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar<br />

disorder (revision). http://www.psych.org/psych_pract/treatg/pg/bipolar_revisebook_1.cfm#a<br />

(Accessed September 20, 2004).<br />

5. Ghaemi SN, Hsu DJ, Soldani F, Goodwin FK. Antidepressants in bipolar disorder: the case<br />

for caution. Bipolar Disord. 2003;5:421-33.<br />

6. Thomson MICROMEDEX. Drug-Reax® Interactive Drug Interactions. MICROMEDEX®<br />

Healthcare Series Vol. 121 expires 9/2004. Accessed September 23, 2004.<br />

7. FDA Public Health Advisory: Worsening depression and suicidality in patients being treated<br />

with antidepressant medications. March 22, 2004.<br />

http://www.fda.gov/cder/drug/antidepressants/AntidepressanstPHA.htm (Accessed<br />

September 23, 2004).<br />

8. FDA Talk Paper. FDA Issues Public Health Advisory on Cautions for Use of Antidepressants<br />

in Adults and Children. March 22, 2004.<br />

http://www.fda.gov/bbs/topics/ANSWERS/2004/ANS01283.html (Accessed September 23,<br />

2004).<br />

9. American Diabetes Association, American Psychiatric Association, American Association of<br />

Clinical Endocrinologists, and the North American Association for the Study of Obesity.<br />

Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes<br />

Care. 2004;27:596-601.<br />

10. MedWatch 2004 Safety Alert: Zyprexa (olanzapine)<br />

http://www.fda.gov/medwatch/SAFETY/2004/zyprexa.htm (Accessed September 23, 2004).<br />

11. Shi L, Namjoshi MA, Swindle R, et al. Effects of olanzapine alone and olanzapine/fluoxetine<br />

combination on health-related quality of life in patients with bipolar depression: Secondary<br />

analyses of a double-blind, placebo-controlled, randomized clinical trial. Clin Ther.<br />

2004;26:125-134.<br />

12. Olanzapine/fluoxetine (Symbyax) for bipolar depression. The Medical Letter. 2004;46:23-24.<br />

13. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind, placebo-controlled study of<br />

lamotrigine monotherapy in outpatients with bipolar I depression: Lamictal 602 Study Group.<br />

J Clin Psychiatry. 1999;60:79-88.<br />

14. Sachs G, Altshuler LL, Ketter TA, et al. Divalproex versus placebo for the treatment of<br />

bipolar depression. Paper presented at: 2001 Annual Meeting of the American College of<br />

Neuropsychopharmacology; December 10,k 2001;Waikoloa, HI.<br />

337


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

New Drug <strong>Pharmacotherapy</strong> <strong>Review</strong>-Pexeva<br />

AHFS 281604<br />

October 27, 2004<br />

I. Overview<br />

Pexeva is a new salt formulation of the selective serotonin reuptake inhibitor, paroxetine.<br />

Previously, all brand and generic formulations of paroxetine have been in the hydrochloride salt<br />

form (e.g., Paxil®, Paxil CR, generics). Pexeva contains the same active moiety, paroxetine,<br />

but in the mesylate salt form. Although paroxetine mesylate is considered to be bioequivalent to<br />

paroxetine hydrochloride, it is not AB rated to paroxetine hydrochloride due to the difference in<br />

salt form and therefore is considered a pharmaceutical alternative to, but not pharmaceutically<br />

equivalent to paroxetine hydrochloride. According to the manufacturer, the FDA did not require<br />

clinical trials for the approval of paroxetine mesylate due to the bridging toxicology and<br />

pharmacokinetic studies conducted by Synthon Pharmaceuticals, combined with existing data and<br />

published literature regarding the safety and efficacy of paroxetine hydrochloride. 1,2 As a<br />

selective serotonin reuptake inhibitor (SSRI), paroxetine is effective in the treatment of a variety<br />

of psychiatric disorders. The SSRI drug class has emerged as the drug class of choice over many<br />

other psychopharmacological treatments due to an improved safety and tolerability profile as<br />

compared to other classes (e.g., tricyclic antidepressants, monoamine oxidase inhibitors, etc.).<br />

AHFS class 281604 was originally reviewed in July 2003, and this review has been included in the<br />

Appendix for reference. This review encompasses all dosage forms and strengths of paroxetine<br />

mesylate.<br />

II.<br />

Evidence Based Medicine and Current Treatment Guidelines<br />

Depression 3-6<br />

Psychotherapy and antidepressant medications are effective in the treatment of depression. More<br />

than 50% of people with major depression experience marked improvement or complete remission<br />

of symptoms when antidepressant therapy is initiated. 3 Initial antidepressant choices used in the<br />

treatment of depression include SSRIs, venlafaxine, mirtazapine, and bupropion. Additional<br />

agents that have shown efficacy include tricyclic antidepressants and monoamine oxidase<br />

inhibitors (usually reserved for non-responders to other therapies). Selective serotonin reuptake<br />

inhibitors are generally considered first-line therapy for the pharmacologic therapy of depression<br />

due to their safety and tolerability as compared to many other antidepressants.<br />

The treatment of clinical depression includes three phases: an acute phase, a continuation phase,<br />

and a maintenance phase. Treatment during the acute phase may last anywhere from 4 to 12<br />

weeks, with a recommended minimum of 6 to 8 weeks. Drug therapy during this phase attempts to<br />

achieve remission or a return to the patient’s baseline level of symptoms and functioning. After<br />

achieving remission, the patient enters the continuation phase of treatment, where the patient is<br />

kept symptom free for the duration of the current episode. Recommended duration of therapy<br />

ranges from 6-12 months. The goal during the maintenance phase is to protect susceptible patients<br />

against new or future major depressive episodes. The treatment length required for maintenance<br />

therapy has not yet been fully determined, and will vary depending on the frequency and severity<br />

of prior major depressive episodes. A maintenance period of 2 to 3 years is appropriate for some<br />

patients with a recurrence, while life-long maintenance therapy may be most appropriate for<br />

patients with recurrent episodes of depression.<br />

338


Panic Disorder 7-9<br />

Panic disorder is estimated to have a lifetime prevalence of 2-4%. It is often accompanied by<br />

comorbid depression. According to practice guidelines developed by the American Psychiatric<br />

Association in 1998, treatment of panic disorder involves psychotherapy and pharmacological<br />

treatment, or a combination of both. Neither psychotherapy nor drug therapy has been proven to<br />

be superior to the other, but the combination of treatment modalities may be more efficacious than<br />

either alone. Recommended pharmacologic therapy involves an antidepressant (preferably an<br />

SSRI) with benzodiazepines utilized for initial rapid control of symptoms when needed.<br />

Antidepressant therapy may take anywhere from 4-12 weeks for full effect of therapy. Initial<br />

dosing of SSRIs is half that of doses used to treat depression, with subsequent dose titration.<br />

Benzodiazepines are used for early symptom control in combination with other pharmacologic<br />

and/or psychotherapeutic modalities. The guidelines recommend continuation of treatment with<br />

antidepressants for 12-18 months before drug discontinuation is attempted. Some patients may<br />

require chronic therapy for control of symptoms. Despite these guidelines recommending<br />

antidepressant therapy as first line therapy for panic disorder, a recent study of anxiety disorders<br />

found that the increase in SSRI use among these patients has been very modest, with<br />

benzodiazepines being the most common class of drugs used to treat this disorder. 7<br />

Obsessive-Compulsive Disorder (OCD) 10-12<br />

OCD affects about 2-3% of the U.S. population. Practice guidelines for OCD were last published<br />

in 1997. Updated practice guidelines for the treatment and management of OCD are expected in<br />

2006 by the American Psychiatric Association. The most effective treatments for OCD include<br />

cognitive-behavioral therapy and drug therapy with SSRIs. Full benefit of treatment may not be<br />

seen for 10-12 weeks, and doses of SSRIs needed may be higher than those typically used for<br />

depression. There is a high rate of relapse after SSRI therapy is discontinued, but this risk may be<br />

decreased with ongoing cognitive-behavioral therapy. Aside from SSRIs, clomipramine and<br />

venlafaxine are alternative agents for the treatment of OCD.<br />

III. Comparative Indications 13-15<br />

Paroxetine mesylate has three FDA-approved indications: Depression, obsessive-compulsive<br />

disorder (OCD), and panic disorder.<br />

Table 1. Indications for Paroxetine Mesylate<br />

Indication<br />

Generalized anxiety disorder (GAD)<br />

Major depressive disorder<br />

Obsessive compulsive disorder (OCD)<br />

Panic disorder<br />

Post-traumatic stress disorder (PTSD)<br />

Premenstrual dysphoric disorder (PMDD)<br />

Social anxiety disorder<br />

Paroxetine mesylate<br />

<br />

<br />

<br />

IV. Pharmacokinetic Parameters 13,16<br />

The efficacy of paroxetine in depression, obsessive-compulsive disorder (OCD), and panic<br />

disorder is thought to be related to the inhibition of serotonin reuptake in the central nervous<br />

system, thereby enhancing serotonergic activity. Paroxetine mesylate is completely absorbed into<br />

the bloodstream after oral administration and is extensively metabolized via oxidation,<br />

methylation, and conjugation. The metabolites have shown no more than 1/50 of the potency of<br />

the parent compound on inhibiting serotonin reuptake and are not considered significant<br />

contributors to drug activity. Paroxetine exhibits nonlinear kinetics with increasing dose and<br />

duration of use due to saturation of one of the metabolizing enzymes, CYP 450 2D 6 . Paroxetine<br />

distributes throughout the body and is excreted primarily as metabolites in the urine (64%) and<br />

feces (36%). Increased plasma concentrations have been observed in elderly patients, and patients<br />

with renal or hepatic impairment may experience two- to four-fold increases in plasma<br />

339


concentrations. Initial dosing needs to be reduced and length of dose titration interval should be<br />

lengthened in these populations. Paroxetine is roughly 95% protein bound with an elimination<br />

half-life of about 15-20 hours, reaching steady-state concentrations in roughly 10-14 days.<br />

V. Drug Interactions 13,16,17<br />

Paroxetine therapy is contraindicated in conjunction with monoamine oxidase inhibitors (MAOIs)<br />

due to the increased risk of serotonin syndrome or neuroleptic malignant syndrome. For patients<br />

switching therapy between these drugs (paroxetine or an MAOI), at least two weeks should elapse<br />

before the other agent is started. The concomitant use of paroxetine and thioridazine is also<br />

contraindicated due to the increased risk of serious cardiac arrhythmias with combined use. There<br />

are numerous additional potential interactions between paroxetine and other drugs listed in the<br />

table below.<br />

Table 2. Drug-Drug Interactions with Paroxetine 17<br />

Interacting Drugs Significance* Mechanism<br />

Sympathomimetics<br />

Amphetamine<br />

Benzphetamine<br />

Dextroamphetamine<br />

Diethylpropion<br />

Methamphetamine<br />

Phendimetrazine<br />

Phenmetrazine<br />

Phentermine<br />

Level 1<br />

(major /<br />

suspected)<br />

Unknown; increased risk of serotonin<br />

syndrome<br />

Selective 5-HT1 receptor<br />

antagonists (“triptans”)<br />

MAO Inhibitors<br />

**CONTRAINDICATED**<br />

Level 1<br />

(major /<br />

suspected)<br />

Level 1<br />

(major /<br />

probable)<br />

Sibutramine Level 1<br />

(major /<br />

suspected)<br />

Risperidone Level 1<br />

(major /<br />

suspected)<br />

NSAIDs Level 2<br />

(moderate /<br />

suspected)<br />

Tricyclic antidepressants<br />

Amitriptyline<br />

Desipramine<br />

Imipramine<br />

Nortriptyline<br />

Level 2<br />

(moderate /<br />

suspected)<br />

Possibly rapid accumulation of serotonin in the<br />

CNS; increased risk of serotonin syndrome<br />

Possibly rapid accumulation of serotonin in the<br />

CNS; increased risk of serotonin syndrome<br />

Additive serotonergic effects; increased risk of<br />

serotonin syndrome<br />

• Possibly rapid accumulation of serotonin<br />

in the CNS; increased risk of serotonin<br />

syndrome<br />

• Inhibition of risperidone metabolism<br />

leading to elevated risperidone plasma<br />

concentrations and risk for adverse events<br />

Unknown; the risk for adverse GI events may<br />

be increased<br />

Paroxetine may inhibit the metabolism of these<br />

TCAs leading to increased risk for TCA<br />

toxicity<br />

340


Table 2 (cont)<br />

Interacting Drugs Significance* Mechanism<br />

Phenothiazines<br />

**THIORIDAZINE<br />

CONTRAINDICATED**<br />

Level 2<br />

(moderate /<br />

probable)<br />

Cyclosporine Level 2<br />

(moderate /<br />

suspected)<br />

Cyproheptadine Level 2<br />

(moderate /<br />

probable)<br />

Propafenone Level 2<br />

(moderate /<br />

suspected)<br />

St. John’s Wort Level 2<br />

(moderate /<br />

suspected)<br />

Zolpidem Level 3<br />

(minor /<br />

suspected)<br />

L-Tryptophan Level 4<br />

(moderate /<br />

possible)<br />

Oxycodone Level 4<br />

(moderate /<br />

possible)<br />

Beta-Blockers<br />

Carvedilol<br />

Metoprolol<br />

Propranolol<br />

Macrolide antibiotics<br />

Clarithromycin<br />

Erythromycin<br />

Troleandomycin<br />

Heparins<br />

Dalteparin<br />

Enoxaparin<br />

Heparin<br />

Tinzaparin<br />

Level 4<br />

(moderate /<br />

possible)<br />

Level 4<br />

(major /<br />

possible)<br />

Level 4<br />

(moderate /<br />

possible)<br />

Cimetidine Level 4<br />

(moderate /<br />

possible)<br />

Metoclopramide Level 4<br />

(major /<br />

possible)<br />

Wafarin Level 4<br />

(moderate /<br />

possible)<br />

Trazodone Level 4<br />

(major /<br />

possible)<br />

Phenytoin Level 4<br />

(moderate /<br />

possible)<br />

Paroxetine may cause decreased phenothiazine<br />

metabolism leading to increased toxicity<br />

including cardiac arrhythmias with thioridazine<br />

SSRIs may inhibit CYP3A4 metabolism<br />

leading to cyclosporine toxicity<br />

Cyproheptadine, a serotonin antagonist, may<br />

lead to decreased antidepressant efficacy of<br />

SSRIs<br />

Paroxetine may inhibit the metabolism of<br />

propafenone leading to increased toxicity<br />

Possible additive serotonin reuptake inhibition<br />

leading to increased sedative-hypnotic effects<br />

Possible inhibition of zolpidem metabolism<br />

leading to increased zolpidem effects<br />

Combination of inhibition of serotonin<br />

reuptake and increased substrate availability<br />

from tryptophan supplementation leading to<br />

toxicity<br />

Unknown; increased risk of serotonin<br />

syndrome<br />

Inhibition of CYP2D6 metabolism of betablockers<br />

leading to increased beta-blocker<br />

effect (bradycardia)<br />

Potential inhibition of SSRI metabolism<br />

leading to increased risk of serotonin syndrome<br />

SSRI-induced impairment of platelet function<br />

leading to increased risk of bleeding with<br />

heparin<br />

Cimetidine may inhibit first pass metabolism<br />

of SSRIs leading to increased SSRI plasma<br />

levels and potential for toxicity<br />

Unknown; potential increased risk, for<br />

serotonin syndrome<br />

Unknown; the anticoagulant effect of warfarin<br />

may be increased (potentially increased<br />

bleeding with unaltered PT)<br />

Inhibition of trazodone metabolism leading to<br />

increased serotonin levels and the risk for<br />

serotonin syndrome<br />

Unknown; serum levels of paroxetine and<br />

phenytoin have been reported, leading to<br />

reduced efficacy<br />

341


Table 2 (cont)<br />

Interacting Drugs Significance* Mechanism<br />

Linezolid Level 4<br />

(major /<br />

possible)<br />

Nefazodone Level 4<br />

(major /<br />

possible)<br />

Tramadol Level 4<br />

(major /<br />

possible)<br />

*Efacts classification (Severity / Documentation)<br />

Possible excessive accumulation of serotonin<br />

leading to serotonin syndrome<br />

Additive or synergistic effects on serotonin;<br />

increased risk of serotonin syndrome<br />

Additive serotonin effects; increased risk of<br />

serotonin syndrome<br />

Additional drug-drug interactions included in the package labeling include phenobarbital<br />

(potentially reduced plasma levels of paroxetine leading to decreased efficacy),<br />

flecainide/encainide (similar to propafenone above), quinidine (inhibition of CYP2D6 enzyme),<br />

procyclidine (increased levels of procyclidine leading to anticholinergic side effects), and<br />

theophylline (increased toxicity).<br />

VI. Adverse Drug Events 13,16<br />

Adverse events seen most commonly with paroxetine use involve the gastrointestinal system,<br />

CNS, sexual dysfunction, sweating, and asthenia. Dose comparison studies in major depressive<br />

disorder have shown an increased incidence of some adverse events with increasing dose, but this<br />

dose dependency was not seen in trials in OCD or panic disorder. Patients may adapt to some<br />

adverse events over a 4-6 week time period (e.g., nausea and dizziness), but other adverse effects<br />

may remain (e.g., asthenia, dry mouth and somnolence).<br />

Paroxetine is not approved for use in patients


Table 3. Most Common Reasons for Paroxetine Discontinuation in Clinical Trials<br />

Major Depressive<br />

Disorder<br />

OCD<br />

Panic Disorder<br />

Adverse Event Paroxetine Placebo Paroxetine Placebo Paroxetine Placebo<br />

CNS<br />

Somnolence 2.3% 0.7% -- 1.9% 0.3%<br />

Insomnia -- -- 1.7% 0% 1.3% 0.3%<br />

Agitation 1.1% 0.5% --<br />

Tremor 1.1% 0.3% --<br />

Dizziness -- -- 1.5% 0%<br />

Gastrointestinal<br />

Constipation -- -- 1.1% 0%<br />

Nausea 3.2% 1.1% 1.9% 0% 3.2% 1.2%<br />

Diarrhea 1.0% 0.3% --<br />

Dry mouth 1.0% 0.3% --<br />

Vomiting 1.0% 0.3% --<br />

Other<br />

Asthenia 1.6% 0.4% 1.9% 0.4%<br />

Abnormal<br />

1.6% 0% 2.1% 0%<br />

ejaculation<br />

Sweating 1.0% 0.3% --<br />

Impotence -- 1.5% 0%<br />

Adapted from “Adverse Reactions: Associated with Discontinuation of Treatment” table, package labeling. 13<br />

The following table lists the most commonly observed adverse events associated with paroxetine<br />

use in clinical trials, where the incidence was >5% and at least twice as common as placebo.<br />

Table 4. Most Commonly Observed Adverse Events in Paroxetine Clinical Trials<br />

Adverse Event Paroxetine Placebo<br />

Major Depressive Disorder 1 (n=421) (n=421)<br />

Nausea 26% 9%<br />

Somnolence 23% 9%<br />

Asthenia 15% 6%<br />

Dizziness 13% 6%<br />

Insomnia 13% 6%<br />

Ejaculatory disturbance 13% 0%<br />

Sweating 11% 2%<br />

Other male genital disorders 10% 0%<br />

Tremor 8% 2%<br />

Decreased appetite 6% 2%<br />

Nervousness 5% 3%<br />

OCD 2 (n=542) (n=265)<br />

Somnolence 24% 7%<br />

Nausea 23% 10%<br />

Abnormal ejaculation 23% 1%<br />

Dry mouth 18% 9%<br />

Constipation 16% 6%<br />

Dizziness 12% 6%<br />

Tremor 11% 1%<br />

Decreased appetite 9% 3%<br />

Sweating 9% 3%<br />

Impotence 8% 1%<br />

343


Table 4 (cont)<br />

Adverse Event Paroxetine Placebo<br />

Panic Disorder 3 (n=469) (n=324)<br />

Abnormal ejaculation 21% 1%<br />

Asthenia 14% 5%<br />

Sweating 14% 6%<br />

Libido decreased 9% 1%<br />

Tremor 9% 1%<br />

Female genital disorders 9% 1%<br />

Decreased appetite 7% 3%<br />

Impotence 5% 0%<br />

1 Duration of trials = 6 weeks; paroxetine dose range = 20-50mg/day<br />

2 Duration of trials = 12 weeks; paroxetine dose range = 20-60mg/day<br />

3 Duration of trials = 10-12 weeks; paroxetine dose range = 10-60mg/day<br />

VII. Dosing and Administration 13,16<br />

Paroxetine mesylate should be administered as a single daily dose with or without food. It is<br />

formulated in 10mg, 20mg, 30mg, and 40mg tablets. The 20mg tablet is scored. If dose increases<br />

are desired, the dose should be increased by 10mg per day at intervals of at least one week.<br />

Table 5. Dosing and Indication for Paroxetine Mesylate<br />

Indication Initial Dose Maximum Dose<br />

Depression 20mg once daily 50mg daily<br />

OCD 20mg once daily 60mg daily<br />

Panic disorder 10mg once daily 60mg daily<br />

Special Dosing Considerations<br />

For elderly or debilitated patients, or those with severe renal or hepatic impairment, paroxetine<br />

mesylate should be initiated at a dose of 10mg once daily and should not exceed 40mg daily.<br />

Adverse effects such as dizziness, sensory disturbances, agitation, anxiety, nausea, and sweating<br />

may be experienced if paroxetine is discontinued too abruptly. When discontinuing treatment<br />

with paroxetine, gradual reduction in dose is recommended to avoid withdrawal-like symptoms.<br />

Recommendations for dose tapering include 10mg/day dose reduction at weekly intervals. When<br />

the patient has received 20mg/day for 1 week, the drug may be stopped.<br />

VIII. Comparative Effectiveness<br />

Efficacy data in the package labeling for paroxetine mesylate is the same as that found in the<br />

package labeling for paroxetine hydrochloride due to the FDA approval process described in the<br />

Overview. According to package labeling, efficacy data for paroxetine was contributed from six<br />

placebo-controlled trials for major depressive disorder, two studies for OCD, and three studies for<br />

panic disorder. Details for these studies are not included in the package labeling. Additional<br />

clinical trials are summarized in the table below.<br />

344


Table 6. Clinical Trials of Paroxetine in Major Depressive Disorder, OCD & Panic Disorder<br />

StudyDesign/ Treatment Endpoints Results<br />

Sample/Duration<br />

Baldwin DS, et.al. 20 Paroxetine 20-<br />

40mg/day<br />

Double blind Nefazodone 200-<br />

600mg/day<br />

N=108<br />

4 months<br />

(Depression)<br />

• HAM-D<br />

• HAM-A<br />

• CGI<br />

• Montgomery-<br />

Ashberg Depression<br />

Rating Scale<br />

• Patient Global<br />

Assessment Scale<br />

• No significant<br />

differences between<br />

treatment regimens in<br />

treatment efficacy or<br />

adverse events<br />

• Both therapies were<br />

efficacious in maintaining<br />

remission during the<br />

continuation phase<br />

Ballenger JC, et.al. 21<br />

Double blind<br />

N=278<br />

10 weeks<br />

(Panic disorder)<br />

Bandelow B, et.al. 22<br />

Double blind<br />

N=225<br />

12 weeks<br />

(Panic disorder)<br />

Paroxetine 10mg,<br />

20mg or 40mg/day<br />

• Sertraline<br />

50-150mg/day<br />

• Paroxetine<br />

40-60mg/day<br />

• Percent of<br />

patients free from<br />

panic attacks<br />

• Change from<br />

baseline in number of<br />

full panic attacks<br />

• Percentage<br />

of patients with 50%<br />

reduction in full panic<br />

attacks<br />

• CGI score<br />

• Panic and<br />

Agoraphobia Scale<br />

(PAS)<br />

• CGI<br />

Significant differences for<br />

40mg dose only compared<br />

to placebo in 3 endpoints:<br />

% patients free from panic<br />

attacks, number of full<br />

panic attacks, and CGI<br />

score (p35% decrease in Y-BOCS<br />

score)<br />

• No difference between<br />

groups in anxiety scale<br />

• Significantly better<br />

improvement in depression<br />

scale at end of study with<br />

paroxetine (p=0.042)<br />

• Both treatments<br />

equally efficacious; similar<br />

rates of adverse events<br />

345


Table 6 (cont)<br />

StudyDesign/<br />

Sample/Duration<br />

Fava M, et.al. 24<br />

Double blind<br />

N=284<br />

10-16 weeks<br />

(Depression)<br />

Kroenke K, et.al. 25<br />

Open label<br />

N=537<br />

9 months<br />

(Depression)<br />

Montgomery S. 26<br />

Meta-analysis<br />

N=1924 (P)<br />

N=141 (C<br />

N=1693 (T)<br />

39 studies<br />

(Depression)<br />

Mundo E, et.al. 27<br />

Single blind<br />

N=30 adults<br />

10 weeks<br />

(OCD)<br />

Treatment Endpoints Results<br />

Fluoxetine 20-<br />

60mg/day<br />

Sertraline 50-<br />

200mg/day<br />

Paroxetine 20-<br />

60mg/day<br />

Fluoxetine 23.4mg<br />

mean dose/day<br />

Sertraline 72.8mg<br />

mean dose/day<br />

Paroxetine 23.5mg<br />

mean dose/day<br />

Paroxetine<br />

Clomipramine<br />

TCAs<br />

Paroxetine 40-<br />

60mg/day<br />

Fluvoxamine 200-<br />

300mg/day<br />

Citalopram 40-<br />

60mg/day<br />

• >50% decrease<br />

or total score 50% decrease or<br />

total score 50% reduction<br />

in HAM-D; 39-48% of<br />

patients met criteria of total<br />

HAM-D score 0.05)<br />

• Paroxetine had<br />

significantly lower<br />

incidence of adverse events<br />

compared to clomipramine<br />

(p=0.02) and other TCAs<br />

(p


Table 6 (cont)<br />

StudyDesign/<br />

Sample/Duration<br />

Wade A, et.al. 28<br />

Double blind<br />

N=197<br />

6 months<br />

(Depression)<br />

Treatment Endpoints Results<br />

Paroxetine 20-<br />

30mg/day<br />

Mirtazapine 30-<br />

45mg/day<br />

• >50% decrease<br />

or total score


X. Recommendations<br />

No brand of paroxetine mesylate (Pexeva) is recommended for preferred status.<br />

348


References<br />

1. Synthon Pharmaceuticals, LTD. Pexeva: frequently asked questions. Accessed via<br />

internet at www.pexeva.com/healthcare/faq.htm on 9/18/2004.<br />

2. FDA Center for Drug Evaluation and Research: Electronic Orange Book: Approved<br />

Drug Products with Therapeutic Equivalence Evaluations. Accessed via internet at<br />

www.fda.gov/cder/orange/default.htm on 9/20/2004.<br />

3. Practice guideline for the treatment of patients with major depressive disorder (revision).<br />

American Psychiatric Association. Am J Psychiatry. 2000 Apr. 157(suppl 4):1-45.<br />

4. Paykel ES. Continuation and maintenance therapy in depression. British Medical<br />

Bulletin. 2001;57:145-159.<br />

5. VHA/DOD clinical practice guideline for the management of major depressive disorder<br />

in adults. Washington (DC): Department of Veterans Affairs (U.S.); 2000.<br />

6. Institute for Clinical Systems Improvement (ICSI). Major depression in adults for mental<br />

health care. Bloomington (MN): Institute for Clinical Systems Improvement (ICSI); 2004<br />

May.<br />

7. Bruce SE, Vasile RG, Goisman RM, et.al. Are benzodiazepines still the medication of<br />

choice for patients with panic disorder with or without agoraphobia Am J Psychiatry<br />

2003;160:1432-1438.<br />

8. American Psychiatric Association. Practice guideline for the treatment of patients with<br />

panic disorder. Am J Psychiatry 1998;155(May supplement).<br />

9. Doyle AD, Pollack MH. Long-term management of panic disorder. J Clin Psychiatry<br />

2004;65(suppl 5):24-28.<br />

10. March JS, Frances A, Kahn DA, Carpenter D, eds. The expert consensus guideline series:<br />

treatment of obsessive-compulsive disorder. J Clin Psychiatry 1997;58 (suppl 4).<br />

Accessed via internet at www.psychguides.com/ocgl.html on 9/22/2004.<br />

11. Jenike MA. Obsessive-compulsive disorder. N Engl J Med 2004;350:259-265.<br />

12. Kaplan A, Hollander E. A review of pharmacologic treatments for obsessive-compulsive<br />

disorder. Psychiatric Services 2003;54:1111-1118.<br />

13. Synthon Pharmaceuticals, LTD. Pexeva package labeling. Chapel Hill, NC. Accessed<br />

9/18/2004 via internet at www.pexeva.com.<br />

14. GlaxoSmithKline. Paxil package labeling, Research Triangle Park, NC. Accessed<br />

9/18/2004 via internet at www.paxil.com.<br />

15. GlaxoSmithKline. Paxil CR package labeling. Research Triangle Park, NC. Accessed<br />

9/18/2004 via internet at www.paxilcr.com.<br />

16. Paroxetine monograph. Clinical Pharmacology, 2004. Accessed September 15, 2004 via<br />

internet at: www.cp.gsm.com.<br />

Wolters Kluwer Health, Inc. EFacts: Drug Interaction Facts. Accessed September 18.<br />

2004 via internet at: www.factsandcomparisons.com.<br />

17. FDA. FDA statement on recommendations of the psychopharmacologic drugs and<br />

pediatric advisory committees 9/16/2004. Accessed September 21, 2004 via internet at<br />

www.fda.gov/bbs/topics/news/2004/NEW01116.html.<br />

18. APA. APA statement on the FDA’s hearing on antidepressant use in pediatric patients<br />

9/16/2004. Accessed 9/21/2004 via internet at www.psych.org.<br />

19. Baldwin DS, Hawley CJ, Mellor’s K. A randomized, double-blind, controlled<br />

comparison of nefazodone and paroxetine in the treatment of depression: safety,<br />

tolerability, and efficacy in the continuation phase treatment. J Psychopharmacol<br />

2001;15(3):161-165.<br />

20. Ballenger JC, Wheadon DE, Steiner M, et.al. Double-blind, fixed-dose, placebocontrolled<br />

study of paroxetine in the treatment of panic disorder. Am J Psychiatry<br />

1998;155:36-42.<br />

21. Bandelow B, Behnke K, Loenoir S, et.al. Sertraline versus paroxetine in the<br />

treatment of panic disorder: an acute, double-blind noninferiority comparison. J Clin<br />

Psychiatry 2004;65(3):405-13.<br />

22. Denys D, van der Wee N, van Megen H, Westenberg H. A double blind comparison<br />

of venlafaxine and paroxetine in obsessive-compulsive disorder. J Clin<br />

Psychopharmacol 2003;23(6):568-575.<br />

349


23. Fava M, Hoog SL, Judge RA, et.al. Acute efficacy of fluoxetine versus sertraline and<br />

paroxetine in major depressive disorder including effects of baseline insomnia. J Clin<br />

Psychopharmacol 2002;22:137-147.<br />

24. Kroenke K, West SL, Swindle R, et.al. Similar effectiveness of paroxetine,<br />

fluoxetine, and sertraline in primary care. JAMA 2001;286:2947-2955.<br />

25. Montgomery SA. A meta-analysis of the efficacy and tolerability of paroxetine<br />

versus tricyclic antidepressants in the treatment of major depression. Int Clin<br />

Pyschopharmacol 2001;16:169-178.<br />

26. Mundo E, Bianchi L, Bellodi L. Efficacy of fluvoxamine, paroxetine, and citalopram<br />

in the treatment of obsessive-compulsive disorder: a single-blind study. J Clin<br />

Psychopharmacol. 1997;17(4):267-71.<br />

27. Wade A, Crawford, GM, Angus M, et.al. A randomized, double-blind, 24 week<br />

study comparing the efficacy and tolerability of mirtazapine and paroxetine in<br />

depressed patients in primary care. Int Clin Psychopharm 2003;18:133-141.<br />

28. Åberg-Wistedt A, Ågren H, Ekselius L, et.al. Sertraline versus paroxetine in major<br />

depression: Clinical outcome after six months of continuous therapy. J Clin<br />

Psychopharmacol 2000;20(6):645-652.<br />

29. Hollander E, Allan A, Steiner M, et.al. Acute and long-term treatment and<br />

prevention of relapse of obsessive-compulsive disorder with paroxetine. J Clin<br />

Psychiatry 2003;64:1113-1121.<br />

30. Doyle A, Pollack MH. Long-term management of panic disorder. J Clin Psychiatry 2004;<br />

65[suppl 5]:24-28.<br />

350


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

New Drug <strong>Pharmacotherapy</strong> <strong>Review</strong>-<br />

Spiriva (Tiotropium)<br />

AHFS Class 120808<br />

October 27, 2004<br />

I. Overview<br />

Tiotropium is a long acting, quaternary ammonium derived from ipratropium. Both agents are<br />

nonselective muscarinic antagonists of M1, M2, and M3 receptors. Although compared to<br />

ipratropium, tiotropium has a 6 to 20 fold greater affinity for these receptors. 1 Additionally, the<br />

muscarinic receptor-drug complex half-lives differ between the two anticholinergics; for<br />

tiotropium the muscarinic receptor-drug complex half lives average 14.6 hours, 3.6 hours, and<br />

34.7 hours for the M1, M2, and M3 receptors, respectively. For ipratropium, the muscarinic<br />

receptor-drug complex half lives average 0.11 hours, 0.035 hours, and 0.26 hours for the M1, M2,<br />

and M3 receptors, respectively. 2<br />

Both agents are bronchodilators used for patients with chronic obstructive pulmonary disease<br />

(COPD), with tiotropium used as once daily therapy compared to the four times daily use of<br />

ipratropium. At the end of one year of treatment, tiotropium improved FEV 1 trough over baseline<br />

by 120 ml while ipratropium had a negative effect on FEV 1 trough , with a decline of 30 ml from<br />

baseline(p


acetylcholine at muscarinic receptors while β 2 -agonists like albuterol and salmeterol stimulate β 2<br />

receptors. The nonbronchodilator effects of these drug classes differ as well. For example,<br />

muscarinic receptors play a role in mucus secretion, while long-acting β 2 -agonists have effects on<br />

mucociliary transport and neutrophils. The potential benefit of combining these two types of<br />

bronchodilators for those with COPD makes sense, but no clinical data are available to fully<br />

support this idea. 5 Of note, while a methylxanthine like theophylline is also considered a weak<br />

bronchodilator, reviewers no longer recommend theophylline as initial therapy. 5,8<br />

Tashkin and Cooper have developed an algorithm that specifically suggests combining tiotropium<br />

with salmeterol or formoterol for those with Stage III or Stage IV COPD; these reviewers are<br />

confident that data demonstrating additive effects of ipratropium and either salmeterol or<br />

formoterol can be extrapolated to combining tiotropium with the latter two agents. 5 In the same<br />

issue of Chest, however, the editorial addressing the Tashkin and Cooper piece does not support<br />

combination long-acting bronchodilator therapy. Although the rationale is compelling given their<br />

complementary mechanisms of bronchodilation, the authors believe it is premature to support<br />

long-acting bronchodilator therapy combination. 7<br />

Table 1. Classification of Chronic Obstructive Pulmonary Disease 4<br />

Stage<br />

Characteristics<br />

0: At Risk<br />

• Normal spirometry<br />

• Chronic symptoms (cough, sputum production)<br />

I: Mild COPD<br />

II: Moderate COPD<br />

III: Severe COPD<br />

IV: Very Severe COPD<br />

• FEV 1 /FVC < 70%<br />

• FEV 1 > 80% predicted<br />

• With or without chronic symptoms (cough,<br />

sputum production)<br />

• FEV 1 /FVC < 70%<br />

• < 50% < FEV 1 < 80% predicted<br />

• With or without chronic symptoms (cough,<br />

sputum production)<br />

• FEV 1 /FVC < 70%<br />

• < 30% < FEV 1 < 50% predicted<br />

• With or without chronic symptoms (cough,<br />

sputum production)<br />

• FEV 1 /FVC < 70%<br />

• FEV 1


Table 2. Chronic Obstructive Pulmonary Disease Therapy by Stage 4<br />

0: At I:Mild II:Moderate III:Severe IV: Very Severe<br />

Risk<br />

Avoidance of Risk Factors, influenza vaccine<br />

Add short-acting bronchodilator when needed<br />

Add regular treatment with one or more long-acting<br />

bronchodilators; Add rehabilitation<br />

Add inhaled corticosteroids if repeated<br />

exacerbations<br />

Add long-term<br />

oxygen if chronic<br />

respiratory failure.<br />

Consider surgical<br />

treatments<br />

Table 3. Use of Bronchodilators in Stable COPD 4<br />

• Bronchodilator medications are central to symptom management in COPD<br />

• Inhaled therapy is preferred.<br />

• The choice between β 2 -agonist, anticholinergic, theophylline, or combination therapy<br />

depends on availability and individual response in terms of symptom relief and side effects.<br />

• Bronchodilators are prescribed on an as-needed or on a regular basis to prevent or reduce<br />

symptoms.<br />

• Long-acting inhaled bronchodilators are more effective and convenient, but more expensive.<br />

• Combining bronchodilators may improve efficacy and decrease the risk of side effects<br />

compared to increasing the dose of a single bronchodilator.<br />

III.<br />

Indications<br />

Ipratropium is indicated for the long-term once daily, maintenance treatment of bronchospasm<br />

associated with COPD, including chronic bronchitis and emphysema. 9<br />

IV.<br />

Pharmacokinetics<br />

Table 4 summarizes the pharmacokinetics of tiotropium. Tiotropium, administered via dry powder<br />

inhalation, is primarily delivered to the GI tract; the rest goes to the lung. 9<br />

Table 4. Pharmacokinetic Parameters of Tiotropium 9<br />

Tmax (minutes) • 5<br />

Absolute Bioavailability • 19.5%<br />

Protein Binding • 72%<br />

Metabolism • Nonenzymatically cleaved to the alcohol N-<br />

methylscopine and dithieneglycolic acid, both<br />

inactive<br />

Elimination • Urinary excretion 14%<br />

• 86% goes through the GI tract and is eliminated<br />

via feces<br />

V. Drug Interactions<br />

None known.<br />

353


VI.<br />

Adverse Drug Events<br />

The adverse events reported with tiotropium, ipratropium, and placebos are summarized in Table<br />

5.<br />

Table 5. Adverse Events with Tiotropium 9<br />

Placebo-controlled trials<br />

Adverse Tiotropium Placebo<br />

Reaction (n=550)<br />

(n=371)<br />

Gastrointest<br />

inal<br />

Ipratropium-controlled trials<br />

Tiotropium Ipratropium<br />

(n=356)<br />

(n=179)<br />

Abdominal<br />

pain<br />

Constipation<br />

Dry mouth<br />

Dyspepsia<br />

Vomiting<br />

5<br />

4<br />

16<br />

6<br />

4<br />

3<br />

2<br />

3<br />

5<br />

2<br />

6<br />

1<br />

12<br />

1<br />

1<br />

6<br />

1<br />

6<br />

1<br />

2<br />

Respiratory<br />

Epistaxis<br />

Pharyngitis<br />

Rhinitis<br />

Sinusitis<br />

Upper<br />

respiratory<br />

tract<br />

infection<br />

4<br />

9<br />

6<br />

11<br />

41<br />

2<br />

7<br />

5<br />

9<br />

37<br />

1<br />

7<br />

3<br />

3<br />

43<br />

1<br />

3<br />

2<br />

2<br />

35<br />

Miscellaneous<br />

Accidents<br />

Chest pain<br />

(nonspecific)<br />

Edema,<br />

dependent<br />

Infection<br />

Moniliasis<br />

Myalgia<br />

Rash<br />

Urinary tract<br />

infection<br />

13<br />

7<br />

5<br />

4<br />

4<br />

4<br />

4<br />

7<br />

11<br />

5<br />

4<br />

3<br />

2<br />

3<br />

2<br />

5<br />

5<br />

3<br />

3<br />

1<br />

3<br />

4<br />

2<br />

4<br />

8<br />

2<br />

5<br />

3<br />

2<br />

3<br />

2<br />

2<br />

VII.<br />

Dosing and Administration<br />

Tiotropium is administered as a single daily 18 µg (one capsule) dose via a HandiHaler ® dry<br />

powder inhaler. It may be administered in the morning, midday, or evening with similar effects. 9<br />

354


VIII. Effectiveness<br />

Tiotropium is one of the few drugs that have come to market in the United States with strong<br />

clinical data available in published form. Table 6 summarizes the results of two placebocontrolled<br />

studies, two comparative studies with ipratropium, and one comparative study vs.<br />

salmeterol.<br />

Two placebo-controlled studies with tiotropium demonstrated that the active treatment was more<br />

effective than placebo in improving lung function and decreasing the amount of rescue medication<br />

in the first study of 13 weeks 10 and more effective than placebo in improving lung function,<br />

decreasing the amount of rescue medication, decreasing the number of exacerbations, decreasing<br />

the number of hospitalizations, and improving quality of life measures (via the St George’s<br />

Respiratory Questionnaire) in a 1 year follow-on study. 11 In both the placebo-controlled studies,<br />

tiotropium had an increased incidence of drug mouth, p


Table 6. Clinical Efficacy Studies for Tiotropium<br />

Treatment (n)<br />

[duration] {Ref}<br />

Tiotropium qd<br />

vs. placebo qd<br />

via dry powder<br />

inhaler (470) [13<br />

wks]{10}<br />

Tiotropium qd<br />

vs. placebo qd<br />

via dry powder<br />

inhaler (921) [12<br />

mo]{11}<br />

Tiotropium qd<br />

vs. ipratropium<br />

qid via metered<br />

dose inhaler<br />

(288) [13<br />

wks]{12}<br />

Tiotropium qd<br />

vs. ipratropium<br />

qid via metered<br />

dose inhaler<br />

(535) [12<br />

mo]{13}<br />

Tiotropium qd<br />

vs. salmeterol<br />

bid and placebo<br />

bid, both by<br />

metered dose<br />

inhaler (623) [6<br />

mo]{15}<br />

Change in Trough<br />

FEV 1 at End of<br />

Study (Liters)<br />

T +0.11, P – 0.04<br />

(p


Stable Therapy: Patients who’s COPD is controlled on ipratropium should not be switched to<br />

tiotropium. As discussed in recent reviews, neither the GOLD guidelines nor clinical data strongly<br />

support a specific drug, combinations of drugs, or the order in which the drugs should be<br />

employed. Therefore, if a patient is well managed on a ‘cocktail’ of ipratropium and other agents<br />

(i.e., β 2 -agonists and/or corticosteroids) there is no clinical reason to change this regimen. 4-6,8<br />

Impact on Physician Visits: Most of the clinical data published to date has included evaluation<br />

of health resources, as measured by fewer hospital visits. Information on potential impact on<br />

physician visits with tiotropium is available from two studies. In the long term comparative study<br />

of tiotropium and ipratropium, the difference between tiotropium and ipratropium for unscheduled<br />

visits was statistically significant, p=0.04. 14 In the comparative study of tiotropium and<br />

salmeterol, unscheduled physician visits were fewer in the tiotropium group compared to the<br />

salmeterol group, but this difference was not statistically significant. 16<br />

IX.<br />

Conclusions<br />

Although tiotropium is chemical derivative of ipratropium, it differs markedly from the current<br />

COPD standard anticholinergic agent. Tiotropium has a 6 to 20 fold greater affinity than<br />

ipratropium for target muscarinic receptors. Where ipratropium must be administered four times<br />

daily, tiotropium is considered a long-acting, once-daily anticholinergic because of the<br />

dramatically different, prolonged muscarinic receptor-drug complex half lives. 1,2<br />

In addition to these chemical and pharmacodynamic differences, the clinical data for tiotropium<br />

demonstrate differences from ipratropium as well. Particularly in the one year comparative study,<br />

the differences between the agents can be seen. Tiotropium improved FEV 1 trough over baseline<br />

by 120 ml while ipratropium had a negative effect on FEV 1 trough, with a decline of 30 ml from<br />

baseline(p


References<br />

1. Haddad EB, Mak JCW, Barnes PJ. Characteristics of [ 3 H]Ba 679 BR, a slowly dissociating<br />

muscarinic antagonist, in human lung: radioligand binding and autoradiographic mapping. Mol<br />

Pharmacol 1994; 45:899-907.<br />

2. Disse B, Speck GA, Rominger KL. Witek TJ, Hammer R. Tiotropium: mechanistical<br />

considerations and clinical profile in obstructive lung disease. Life Sci 1999; 64:457-464.<br />

3. Vincken W, van Noord JA, Greefhorst APM, et al. Improved health outcomes in patients with<br />

COPD during 1 year’s treatment with tiotropium. Eur Resp J 2002;19:209- 216.<br />

4. Global Initiative for Chronic Obstructive Lung Disease, World Health Organization, National<br />

Heart, Lung and Blood Institute. Global strategy for the diagnosis, management, and prevention<br />

of chronic obstructive pulmonary disease. Bethesda (MD): Global Initiative for Chronic<br />

Obstructive Lung Disease, World Health Organization, National Heart, Lung and Blood Institute;<br />

2001. Executive Summary.Updated 2004. url:// www.goldcopd.com Accessed: September 15,<br />

2004<br />

5. Tashkin DP, Cooper CB. The role of long-acting bronchodilators in the management of stabel<br />

COPD. Chest 2004; 125:249-259<br />

6. Sin DD, McAlister FA, Man SFP, Anthonisen NR. Contemporary management of chronic<br />

obstructive pulmonary disease: Scientific review. JAMA 2003; 290:2301-2312.<br />

7. Cazzola M, Matera MG. Long-acting bronchodilators are the first choice option for the treatment<br />

of stable COPD. Chest 2004; 125:9-11.<br />

8. Faulkner MA, Hilleman DE. Pharmacologic treatment of chronic obstructive pulmonary disease:<br />

past, present, and future. <strong>Pharmacotherapy</strong> 2003; 23:1300-1315.<br />

9. Product information for Spiriva HandiHaler ® . Boehringer Ingelheim Pharmaceuticals Inc.<br />

Ridgefield, CT. January 2004.<br />

10. Casaburi R, Briggs DD, Donohue JF, et al. The spirometric efficacy of once-daily dosing with<br />

tiotropium in stable COPD. Chest 2000 118:1294-1302.<br />

11. Casaburi R, Mahler DA, Jones A, et al. A long-term evaluation of once-daily tiotropium in<br />

chronic obstructive pulmonary disease. Eur Resp J 2002;19:217-224.<br />

12. van Noord JA, Bantje TA, Eland ME, Korducki L, Cornelissen PJ.A randomised controlled<br />

comparison of tiotropium and ipratropium in the treatment of chronic obstructive pulmonary<br />

disease. The Dutch Tiotropium Study Group. Thorax. 2000; 55:289-294.<br />

13. Vincken W, van Noord JA, Greefhorst APM, et al. Improved health outcomes in patients with<br />

COPD during 1 year’s treatment with tiotropium. Eur Resp J 2002;19:209- 216.<br />

14. Oosterbrink JB, Rutten-van Molen MPMH, Van Noord JA, Vinken W. One year costeffectiveness<br />

of tiotropium versus ipratropium to treat chronic obstructive pulmonary disease. Eur<br />

Respir J. 2004; 23:241-249.<br />

15. Donohue JF, van Noord JA, Bateman ED, et al. A 6-month, placebo-controlled study comparing<br />

lung function and health status changes in COPD patients treated with Tiotropium or salmeterol.<br />

Chest. 2002; 122:47-55.\<br />

16. Brusaasco V, Hodder R, Miravitlles M, Korducki L, Towse L, Kesten S. Health outcomes<br />

following treatment for six month with once daily tiotropium compared with twice daily<br />

salmeterol in patients with COPD. Thorax 2003; 58:399-404.<br />

358


Alabama Medicaid Agency<br />

Pharmacy and Therapeutics Committee Meeting<br />

New Drug <strong>Pharmacotherapy</strong> <strong>Review</strong> - Caduet<br />

AHFS Class 240608<br />

October 27, 2004<br />

I. Overview<br />

Caduet ® is a combination of two drugs: atorvastatin, a selective, competitive inhibitor of HMG-<br />

CoA reductase (statin, cholesterol-lowering agent); and amlodipine, a long-acting dihydropyridine<br />

calcium antagonist (calcium channel blocker, antihypertensive/antianginal agent). 1<br />

Atorvastatin, which is also available as the brand product Lipitor ® , inhibits HMG-CoA reductase,<br />

the rate limiting enzyme that converts 3-hydroxy-3-methylglutary-coenzyme A to mevalonate, a<br />

precursor of sterols (including cholesterol).<br />

Amlodipine is also available as the brand product Norvasc ® . Amlodipine inhibits the<br />

transmembrane influx of calcium ions into vascular smooth muscle and cardiac muscle, which the<br />

contractile processes are dependent upon.<br />

Both hyperlipidemia and hypertension are risk factors for the development of coronary heart<br />

disease (CHD). 2,3 Therefore, appropriate cholesterol and blood pressure control is essential to<br />

reduce the risk of morbidity and mortality associated with CHD. Atorvastatin/amlodipine<br />

(Caduet ® ) is the first combination product combining both a cholesterol-lowering agent and an<br />

antihypertensive/antianginal agent. Neither atorvastatin nor amlodipine are available generically. 4<br />

There are various combination strengths available for atorvastatin/amlodipine. 1 This review<br />

encompasses all atorvastatin/amlodipine dosage forms and strengths. The HMG-CoA Reductase<br />

Inhibitor (AHFS 240608) pharmacotherapy review in full is available for reference in Appendix<br />

1.<br />

II.<br />

Current Treatment Guidelines<br />

Hyperlipidemia 2,3<br />

The National Cholesterol Education Program (NCEP) Adult Treatment Panel (ATP) has issued<br />

recommendations for cholesterol management. According to the ATP III guidelines, therapy with<br />

lipid-altering agents is one of several components of multiple-risk-factor intervention in<br />

individuals at increased risk for CHD due to hypercholesterolemia. Therapeutic lifestyle changes<br />

(TLC) and drug therapy are the two major treatment modalities. The TLC Diet stresses reductions<br />

in saturated fat and cholesterol intake. The following table defines LDL-C goals and cutpoints for<br />

initiation of TLC and for drug consideration based on the updated ATP III guidelines, 3 issued July<br />

13, 2004. These updates advise physicians to consider new, more intensive treatment options for<br />

people at high and moderately high risk for a heat attack. The new guidelines are endorsed by the<br />

National Heart, Lung, and Blood Institute (NHLBI), the American College of Cardiology, and the<br />

American Heart Association.<br />

359


Table 1. LDL-C Cholesterol Goals and Cutpoints for Therapeutic Lifestyle Changes (TLC) and Drug<br />

Therapy in Different Risk Categories Based on Updated ATP III Guidelines 3<br />

LDL Level at<br />

LDL Goal Which to Initiate LDL Level at Which to Consider<br />

Risk Category<br />

High-Risk: CHD or CHD Risk<br />

Equivalents*<br />

(10-year risk >20%)<br />

Moderately-High Risk: 2+<br />

Risk Factors†<br />

(10-year risk 10-20%)<br />

Moderate-Risk: 2+ Risk<br />

Factors†<br />

(10-year risk


NEW GOALS:<br />

• For high-risk patients, the update lowers the threshold for drug therapy to an LDL-C of<br />

100mg/dL or higher and recommends drug therapy for those high-risk patients whose LDL-C<br />

is 100 to 129mg/dL. Previously this threshold for drug therapy was an LDL-C of 130mg/dL,<br />

and made drug treatment optional for LDL-C of 100 to 129mg/dL.<br />

• For moderately high-risk patients, the goal remains an LDL-C


Angina 7<br />

The American College of Cardiology/American Heart Association Task Force for the<br />

Management of Patients with Chronic Stable Angina has issued treatment guidelines.<br />

Recommendations to prevent Myocardial Infarction and Death and to Reduce Angina Symptoms<br />

include the following pharmacologic therapy:<br />

• Aspirin in the absence of contraindication. Evidence also suggests low-intensity<br />

anticoagulation with warfarin in addition to aspirin.<br />

—If aspirin is absolutely contraindicated, clopidogrel.<br />

• Beta-blockers as initial therapy in the absence of contraindications in patients with or without<br />

prior MI. Evidence also suggests long-acting nondihydropyridine calcium antagonists instead<br />

of beta-blockers as initial therapy:<br />

—If beta-blockers are contraindicated or initial beta-blocker therapy is not effective, calcium<br />

antagonists (short-acting, dihydropyridines should be avoided) should then be<br />

administered.<br />

—If beta-blockers are unacceptable due to side effects, calcium channel-antagonists<br />

(short-acting, dihydropyridines should be avoided) or long-acting nitrates may be a<br />

substitute. No conclusive evidence exists to indicate that either long-acting nitrates or<br />

calcium antagonists are superior for long-term treatment for symptomatic relief of angina,<br />

however, the guidelines say that long-acting calcium antagonists are often preferable to<br />

long-acting nitrates for maintenance therapy because of their sustained 24-hour effects.<br />

• Angiotensin converting enzyme inhibitor (ACEI) in all patients with coronary artery disease<br />

who also have diabetes and/or left ventricular systolic dysfunction. Evidence also suggests<br />

use in CAD or other vascular disease.<br />

• LDL-C lowering therapy in subjects with documented or suspected CAD and LDL-C<br />

>130mg/dL, with a target


—There is a positive family history of premature cardiovascular disease, or<br />

—Two or more other CVD risk factors are present.<br />

Amlodipine<br />

14. Hypertension: Amlodipine is indicated for the treatment of hypertension. It may be used<br />

alone or in combination with other antihypertensive agents;<br />

15. Chronic Stable Angina: Amlodipine is indicated for the treatment of chronic stable angina,<br />

for the treatment of confirmed or suspected vasospastic angina and for the treatment of<br />

hypertension. Amlodipine may be used alone or in combination with other antianginal or<br />

antihypertensive agents;<br />

16. Vasospastic Angina (Prinzmetal's or Variant Angina): Amlodipine is indicated for the<br />

treatment of confirmed or suspected vasospastic angina. Amlodipine may be used as<br />

monotherapy or in combination with other antianginal drugs.<br />

*Note the indications for atorvastatin/amlodipine are exactly the same as for its individual components, atorvastatin and<br />

amlodipine (Lipitor ® and Norvasc ® , respectively). 8,9<br />

IV. Pharmacokinetic Parameters 1<br />

Peak concentrations for amlodipine and atorvastatin are achieved at 6 to 12 hours and 1 to 2 hours<br />

post-dosing, respectively. The rate and extent (bioavailability) of atorvastatin and amlodipine<br />

from atorvastatin/amlodipine are not significantly different from the bioavailability of each<br />

medication administered separately. As noted with atorvastatin administration separately,<br />

administration of atorvastatin/amlodipine with food reduces the rate and extent of absorption of<br />

atorvastatin by approximately 32% and 11%, respectively. However, LDL-C reduction is similar<br />

whether atorvastatin is given with or without food. Pharmacokinetic parameters for atorvastatin<br />

and amlodipine following separate administration are provided in the following table.<br />

Table 3. Pharmacokinetic Parameters of Atorvastatin and Amlodipine Following Lipitor ® and<br />

Norvasc ® Administration Separately 1<br />

Parameter Atorvastatin Amlodipine<br />

Absolute Bioavailability 14% 64-90%<br />

T max 1-2 hours 6-12 hours<br />

Steady State Levels<br />

achieved by Day<br />

7-8<br />

Plasma Protein Binding ≥98% 93%<br />

Food Effect on<br />

None<br />

Bioavailability<br />

Metabolism<br />

Excretion<br />

T 1/2<br />

Although rate of absorption is<br />

decreased by 25% and extent of<br />

absorption is decreased by 9%, LDL-<br />

C reduction is similar if given with or<br />

without food.<br />

Extensively metabolized to orthoand<br />

parahydroxylated derivatives and<br />

various beta-oxidation products<br />

which in vitro contribute to 70% of<br />

circulating HMG-CoA inhibitory<br />

activity. In vitro suggests metabolism<br />

via CYP 3A4.<br />

Parent compound and metabolites<br />

primarily excreted in bile, but no<br />

enterohepatic recirculation and


Special Pharmacokinetic Considerations:<br />

Elderly patients: Amlodipine clearance is reduced (AUC increased 40-60%). Thus, a lower initial<br />

amlodipine dose may be required. Although, atorvastatin concentrations are higher in elderly<br />

(40% for C max and 30% for AUC), clinical data suggests a greater degree of LDL-lowering at any<br />

dose in this population compared to younger subjects.<br />

Renal Impairment: No effect on either atorvastatin or amlodipine concentrations. Thus, dose<br />

adjustment is not necessary. Hemodialysis is not expected to effect concentrations of either drug<br />

due to extensive binding to plasma proteins.<br />

Hepatic Impairment: Amlodipine concentrations are increased (AUC 40-60%). Thus, a lower<br />

initial dose may be required. Atorvastatin concentrations are markedly increased in chronic<br />

alcoholic liver disease and by at least 4-fold for both C max and AUC in Childs-Pugh A disease.<br />

C max and AUC are increased by 16- and 11-fold, respectively, in Childs-Pugh B disease. Due to<br />

these atorvastatin increases, atorvastatin/amlodipine is contraindicated in patients with active liver<br />

disease or unexplained persistent elevations in transaminases.<br />

V. Drug Interactions<br />

No drug interaction studies have been conducted with the combination product<br />

atorvastatin/amlodipine; thus, the following information has been provided regarding drug<br />

interactions of atorvastatin and amlodipine administered separately. 1<br />

Due to the atorvastatin component, there is a warning when atorvastatin/amlodipine is used with<br />

certain medications. Since there is an increased risk for myopathy and rhabdomyolysis when<br />

coadministered with cyclosporine, fibric acid derivatives, erythromycin, niacin, or azole<br />

antifungals, use cautiously. Dosage reduction of atorvastatin and monitoring for side effects is<br />

warranted to properly manage these interactions. Specific clinically significant (level 1 [major]<br />

and level 2 [moderate]) drug interactions with atorvastatin/amlodipine are listed in the table<br />

below. 10<br />

364


Table 4. Level 1 Drug Interactions with Atorvastatin/Amlodipine ®10<br />

Drug Significance Mechanism Management<br />

Macrolide<br />

Antibiotics<br />

(azithromycin,<br />

clarithromycin,<br />

erythromycin)<br />

Level 1 (Delayed,<br />

Major, Probable)<br />

Severe myopathy/rhabdomyolysis may occur due<br />

to suspected increased atorvastatin<br />

concentrations due CYP3A4 inhibition. A PK<br />

study determined that atorvastatin concentrations<br />

increased ~40% during coadministration with<br />

Suspend statin until antibiotic therapy complete<br />

or administer alternative statin therapy such as<br />

fluvastatin and pravastatin which are not<br />

metabolized by CYP3A4.<br />

Cyclosporine<br />

Gemfibrozil<br />

Nefazadone<br />

Protease<br />

Inhibitors<br />

(amprenavir,<br />

indinavir,<br />

lopinavir/ritonavir,<br />

nelfinavir,<br />

ritonavir,<br />

saquinavir)<br />

Bile Acid<br />

Sequestrants<br />

(cholestyramine,<br />

colestipol)<br />

Nondihydropyricine<br />

Calcium<br />

Channel<br />

Blockers<br />

(diltiazem,<br />

verapamil)<br />

Azole<br />

Antifungals<br />

(fluconazole,<br />

itraconazole,<br />

ketoconazole,<br />

voriconazole)<br />

Grapefruit Juice<br />

Rifamycins<br />

(rifampin,<br />

rifabutin)<br />

Level 1 (Delayed,<br />

Major, Probable)<br />

Level 1 (Delayed,<br />

Major, Suspected)<br />

Level 1 (Delayed,<br />

Major, Suspected)<br />

Level 2 (Delayed,<br />

Moderate,<br />

Suspected)<br />

Level 2 (Delayed,<br />

Moderate,<br />

Suspected)<br />

Level 2 (Delayed,<br />

Moderate,<br />

Probable)<br />

Level 2 (Rapid,<br />

Moderate,<br />

Probable)<br />

Level 2 (Moderate,<br />

Suspected)<br />

Level 2 (Delayed,<br />

Moderate,<br />

Suspected)<br />

erythromycin.<br />

Myolysis and rhabdomyolysis may occur due to<br />

increased plasma levels and side effects of statins<br />

due to a suspected decrease in metabolism.<br />

Severe myopathy/rhadomyolysis may occur and<br />

the mechanism is unknown.<br />

The risk of statin-induced rhabdomyolysis and<br />

myositis may be increased with certain agents in<br />

this class due to possible nefazodone inhibition<br />

of metabolism by CYP3A4.<br />

Atorvastatin levels may be elevated, increasing<br />

the risk of side effects (eg, rhabdomyolysis) due<br />

to suspected inhibition of atorvastatin first-pass<br />

metabolism CYP3A4.<br />

Pharmacologic effects of atorvastatin may be<br />

reducted due to reduced GI absorption. Note<br />

clinical study found that although atorvastatin<br />

levels were reduced by ~25% during<br />

coadministration with colestipol, LDL-C<br />

reduction was greater when the agents were<br />

coadministered versus given alone.<br />

Plasma concentrations of certain statins may be<br />

elevated, increasing the risk of toxicity (eg,<br />

rhabdomyolysis, myositis) possibly due to<br />

inhibition of first-pass metabolism (CYP3A4).<br />

Note a PK study determined steady state digoxin<br />

concentrations were increased by ~20%.<br />

Increased plasma levels and side effects (eg,<br />

rhabdomyolysis) of statins may occur due to<br />

inhibition of first-pass hepatic metabolism of<br />

CYP3A4.<br />

Increased serum levels and side effects (eg,<br />

rhabdomyolysis) of certain statins due to<br />

inhibition of first-pass metabolism (CYP3A4) in<br />

the small intestine.<br />

Statin levels may be reduced, decreasing the<br />

pharmacologic effects due to induction of firstpass<br />

metabolism (CYP3A4) in the intestine and<br />

liver.<br />

If coadministration cannot be avoided, monitor<br />

patients more closely for side effects and<br />

consider reducing the dose of atorvastatin while<br />

carefully monitoring the response.<br />

Avoid statin therapy unless benefits outweigh<br />

the risks.<br />

If coadministration cannot be avoided, monitor<br />

patients more closely for side effects or avoid<br />

statin therapy unless benefits outweigh the risks<br />

Monitor patients more closely for side effects or<br />

avoid statin therapy unless benefits outweigh the<br />

risks.<br />

Separate administration times by as much as<br />

possible (at least 4 hr before the statin),<br />

preferably giving the bile acid sequestrant<br />

before meals.<br />

If coadministration of these agents cannot be<br />

avoided, monitor patients more closely for side<br />

effects and possibly reduce statin dose. Digoxin<br />

levels should be monitored during atorvastatin<br />

therapy and reductions in dosage may be<br />

needed.<br />

If coadministration cannot be avoided, consider<br />

reducing the statin dose and carefully monitor<br />

the patient's response.<br />

Avoid coadministration with grapefruit<br />

products.<br />

Monitor the clinical response of the patient; if<br />

an interaction is suspected, it may be necessary<br />

to administer alternative therapy. Increase statin<br />

dose if needed.<br />

Most of the drug interactions listed above can be managed with appropriate dosing modifications<br />

and monitoring. Atorvastatin should not be used concomitantly with nefazodone and protease<br />

inhibitors unless the benefits of therapy outweigh the risks for potential side effects. However,<br />

nefazodone and protease inhibitors are used in specific patient populations and not the general<br />

population, for which we are evaluating the use of atorvastatin/amlodipine. When considering the<br />

general population, use of any statin would not be precluded due to potentially harmful drug<br />

interactions.<br />

365


VI. Adverse Drug Events 1<br />

Atorvastatin/amlodipine has been evaluated for safety in 1,092 patients in double-blind, placebo<br />

controlled trials in patients with co-morbid hypertension and dyslipidemia.<br />

Atorvastatin/amlodipine is generally well-tolerated, and most adverse events have been mild or<br />

moderate in severity with no peculiar events specific to the combination. Adverse events are<br />

similar in terms of nature, severity, and frequency to those reported with separate administration of<br />

atorvastatin and amlodipine.<br />

Atorvastatin<br />

Of 2,502 patients treated with atorvastatin during clinical trials, 3 times the upper limit of normal [ULN] occurring on two or more<br />

occasions) in serum transaminases occurred in 0.7% of patients who received atorvastatin in<br />

clinical trials. The incidence of these abnormalities is dose dependent with 0.2%, 0.2%, 0.6%, and<br />

2.3% for 10, 20, 40, and 80mg tablets, respectively. These changes generally occurred within the<br />

first three months of treatment. Upon dose reduction, drug interruption, or discontinuation,<br />

transaminase levels return to or near pretreatment levels without sequelae. Due to this adverse<br />

event, liver function tests should be performed prior to and at 12 weeks following both initiation<br />

of therapy and any increase in dosage, and periodically (e.g., semiannually) thereafter. If<br />

increases in transaminases occur, patients should be monitored until the abnormalities resolve. If<br />

an increase in ALT or AST of >3 times ULN persists, a reduction in dose or discontinuation of<br />

atorvastatin/amlodipine is recommended.<br />

Skeletal Muscle Abnormalities<br />

Similar to other HMG-CoA reductase inhibitors, rare cases of rhabdomyolysis with acute renal<br />

failure secondary to myoglobinuria have been reported. In addition to rhabdomyolysis, myalgia<br />

has been reported. Myopathy, defined as muscle aches or muscle weakness in conjunction with<br />

increases in creatinine phosphokinase (CPK) values >10 times ULN, should be considered in any<br />

patient with diffuse myalgias, muscle tenderness or weakness, and/or marked elevation of CPK.<br />

Patients should be advised to promptly report unexplained muscle pain, tenderness or weakness,<br />

particularly if accompanied by malaise or fever. Atorvastatin/amlodipine should be discontinued<br />

if markedly elevated CPK levels occur, or if myopathy is diagnosed or suspected. This risk is<br />

increased during concomitant administration with cyclosporine, fibric acid derivatives,<br />

erythromycin, niacin, and azole antifungals. If coadministration benefits outweigh these risks,<br />

patients should be carefully monitored and periodic CPK determinations may be considered.<br />

Atorvastatin/amlodipine therapy should be temporarily withheld or discontinued in any patient<br />

with an acute, serious condition suggestive of a myopathy or having a risk factor predisposing to<br />

the development of renal failure secondary to rhabdomyolysis.<br />

Adverse events reported in ≥2% of subjects regardless of causality during atorvastatin clinical<br />

trials are listed in the following table.<br />

366


Table 5. Atorvastatin Adverse Events Reported in ≥2% of Subjects During Clinical Trials* 1<br />

Placebo<br />

n=270<br />

10mg<br />

n=863<br />

Atorvastatin Dosage<br />

20mg 40mg<br />

n=36 n=79<br />

80mg<br />

n=94<br />

Body System/Adverse Event<br />

Body as a Whole<br />

Infection<br />

10.0 10.3 2.8 10.1 7.4<br />

Headache<br />

7.0<br />

5.4 16.7 2.5 6.4<br />

Accidental Injury<br />

3.7<br />

4.2<br />

0.0<br />

1.3 3.2<br />

Flu Syndrome<br />

1.9<br />

2.2<br />

0.0<br />

2.5 3.2<br />

Abdominal Pain<br />

0.7<br />

2.8<br />

0.0<br />

3.8 2.1<br />

Back Pain<br />

3.0<br />

2.8<br />

0.0<br />

3.8 1.1<br />

Allergic Reaction<br />

2.6<br />

0.9<br />

2.8<br />

1.3 0.0<br />

Asthenia<br />

1.9<br />

2.2<br />

0.0<br />

3.8 0.0<br />

Digestive System<br />

Constipation<br />

1.8<br />

2.1<br />

0.0<br />

2.5 1.1<br />

Diarrhea<br />

1.5<br />

2.7<br />

0.0<br />

3.8 5.3<br />

Dyspepsia<br />

4.1<br />

2.3<br />

2.8<br />

1.3 2.1<br />

Flatulence<br />

3.3<br />

2.1<br />

2.8<br />

1.3 1.1<br />

Respiratory System<br />

Sinusitis<br />

2.6<br />

2.8<br />

0.0<br />

2.5 6.4<br />

Pharyngitis<br />

1.5<br />

2.5<br />

0.0<br />

1.3 2.1<br />

Skin and Appendages<br />

Rash 0.7 3.9 2.8 3.8 1.1<br />

Musculoskeletal System<br />

Arthralgia<br />

Myalgia<br />

1.5<br />

1.1<br />

*Includes adverse events regardless of causality.<br />

2.0<br />

3.2<br />

Amlodipine<br />

Of over 11,000 patients treated with amlodipine during clinical trials, discontinuation due to<br />

adverse events was required in ~1.5% of patients, which was not significantly different than<br />

placebo (~1%). Most adverse events were of mild or moderate severity, with headache and edema<br />

being the most common (edema is dose dependent as described in following table).<br />

0.0<br />

5.6<br />

5.1<br />

1.3<br />

0.0<br />

0.0<br />

367


Table 6. Amlodipine Adverse Events During Clinical Trials 1<br />

Dose-Related Amlodpine Adverse Events<br />

Amlodipine<br />

2.5mg<br />

5mg<br />

10mg<br />

Placebo<br />

n=520<br />

Adverse Event n=275 n=296<br />

n=268<br />

Edema 1.8 3.0 10.8 0.6<br />

Dizziness 1.1 3.4 3.4 1.5<br />

Flushing 0.7 1.4 2.6 0.0<br />

Palpitation 0.7 1.4 4.5 0.6<br />

Other Amlodipine Adverse Events in >1.0% Subjects in Placebo-Controlled Trials<br />

Amlodipine (%)<br />

Adverse Event<br />

n=1,730<br />

Headache 7.3 7.8<br />

Fatigue 4.5 2.8<br />

Nausea 2.9 1.9<br />

Abdominal Pain 1.6 0.3<br />

Somnolence 1.4 0.6<br />

Gender-Related Differences in Dose-Related Adverse Events<br />

Placebo (%)<br />

n=1,250<br />

Amlodpine<br />

Placebo<br />

Adverse Event<br />

Male %<br />

n=1,218<br />

Female %<br />

N=512<br />

Male %<br />

n=914<br />

Edema 5.6 14.6 1.4 5.1<br />

Flushing 1.5 4.5 0.3 0.9<br />

Palpitations 1.4 3.3 0.9 0.9<br />

Somnolence 1.3 1.6 0.8 0.3<br />

VII. Dosing and Administration 1<br />

Female %<br />

n=336<br />

General Dosing Considerations<br />

Atorvastatin/amlodipine can be used in several different situations:<br />

1. Substitution for atorvastatin and amlodipine following appropriate dose titration of the<br />

medications separately with the equivalent atorvastatin/amlodipine tablet.<br />

2. Substitution with increased amounts of atorvastatin, amlodipine, or both for additional lipid<br />

lowering, antianginal, or blood pressure lowering effects.<br />

3. As initial therapy for one indication and continuation of treatment of the other. The<br />

recommended starting dose should be selected based on the continuation of the component<br />

being used and the recommended starting dose for the added new therapy.<br />

4. As initial treatment for hyperlipidemia and either hypertension or angina. The recommended<br />

starting dose should be based on the appropriate combination of recommendations for the<br />

initial components.<br />

The maximum dose of atorvastatin/amlodipine is 10mg for the amlodipine component and 80mg<br />

for the atorvastatin component, administered once daily. A standard cholesterol-lowering diet<br />

should be initiated before starting atorvastatin therapy, and the patient should continue on this diet<br />

during atorvastatin/amlodipine therapy. Atorvastatin/amlodipine can be administered as a single<br />

dose at any time of day, with or without food.<br />

368


Available Dosage Strengths:<br />

• Amlodipine/atorvastatin 5/10mg<br />

• Amlodipine/atorvastatin 5/20mg<br />

• Amlodipine/atorvastatin 5/40mg<br />

• Amlodipine/atorvastatin 5/80mg<br />

• Amlodipine/atorvastatin 10/10mg<br />

• Amlodipine/atorvastatin 10/20mg<br />

• Amlodipine/atorvastatin 10/40mg<br />

• Amlodipine/atorvastatin 10/80mg<br />

Note: An atorvastatin/amlodipine formulation containing amlodpine 2.5mg is not available,<br />

and this amlodipine dose is recommended in specific situations as described below.<br />

Atorvastatin Dosing<br />

Adults:<br />

The recommended starting dose for Hypercholesterolemia (Heterozygous Familial and<br />

Nonfamilial) and Mixed Dyslipidemia (Fredrickson Types IIa and IIb) is 10 or 20mg once<br />

daily.<br />

• Initiation with a higher dose (e.g., 40mg) may be considered for patients requiring large<br />

LDL-C reductions (e.g., >45%).<br />

• Lipid levels should be evaluated within 2 to 4 weeks after initiation and/or upon titration,<br />

and dose adjusted accordingly.<br />

• LDL-C levels should be used to initiate and evaluate response.<br />

Dosage in Homozygous Familial Hypercholesterolemia is 10 to 80mg daily and should be<br />

used in adjunct to other lipid-lowering therapy (e.g., LDL apheresis).<br />

Pediatrics (10-17 years):<br />

The recommended starting dose is 10mg daily with a maximum of 20mg daily. Doses >20mg<br />

have not been evaluated in this population.<br />

• Dosage adjustments may be made at intervals of 4 weeks or more.<br />

Amlodipine Dosing<br />

Adults:<br />

Initial antihypertensive dose is 5mg once daily with a maximum dose of 10mg once daily.<br />

• Elderly and/or small, fragile individuals should be started on 2.5mg once daily. This<br />

dose may also be used when adding amlodipine to other antihypertensive therapy.<br />

• Dose adjustment should proceed over 7 to 14 days; however, titration may occur more<br />

rapidly if clinically warranted and the patient is assessed frequently.<br />

Initial recommended dose for chronic stable or vasospastic angina is 5-10mg once daily.<br />

• The lower dosage of 5mg is suggested in the elderly and in patients with hepatic<br />

insufficiency.<br />

• Most patients require 10mg for clinical effect.<br />

Pediatrics (6-17 years):<br />

Recommended dose is 2.5 to 5mg once daily. Doses >5mg have not been studied.<br />

Special Considerations for Use<br />

• Contraindicated in patients with active liver disease or unexplained elevations of serum<br />

transaminases.<br />

• Contraindicated in patients with a known hypersensitivity to any component of the<br />

medication.<br />

• Pregnancy Category X. Contraindicated in Pregnancy and Lactation.<br />

• Dose adjustment not needed in renal impairment. Hemodialysis not expected to<br />

significantly enhance clearance of either component.<br />

• Atorvastatin/amlodipine formulation containing 2.5mg of amlodipine is unavailable.<br />

369


VIII. Effectiveness<br />

RESPOND Study 1<br />

A double-blind, placebo controlled study evaluated the safety and efficacy of<br />

atorvastatin/amlodipine (Caduet ® ) in 1,660 patients with co-morbid hypertension and<br />

dyslipidemia. Patients were treated with various fixed doses of atorvastatin/amlodipine (e.g., 5/10,<br />

10/10, 5/20, 10/20, 5/40, 10/40, 5/80, or 10/80), amlodipine alone (5 or 10mg), atorvastatin alone<br />

(10, 20, 40, or 80mg), or placebo. All medication was administered once daily. Additional risk<br />

factors in this population other than hypertension and dyslipidemia included diabetes mellitus<br />

(15%), smokers (22%), and a positive family history of cardiovascular disease (14%). Following<br />

eight weeks of therapy, all eight atorvastatin/amlodipine combination treatment groups<br />

demonstrated statistically significant dose-related reduction in systolic and diastolic blood<br />

pressure (SBP and DBP, respectively), and LDL-C compared to placebo. There was no overall<br />

modification on the effect of either component on SBP, DBP, and LDL-C.<br />

Table 7. Efficacy of Combined Treatments in Reducing Systolic Blood Pressure 1<br />

Parameter/Analysis<br />

AML 0mg Mean Change (mmHg)<br />

(Placebo)<br />

Difference versus<br />

Placebo (mmHg)<br />

AML 5mg Mean Change (mmHg)<br />

ATO 0mg<br />

(Placebo)<br />

-3.0<br />

-<br />

-12.8<br />

ATO<br />

10mg<br />

-4.5<br />

-1.5<br />

-13.7<br />

ATO<br />

20mg<br />

-6.2<br />

-3.2<br />

-15.3<br />

ATO<br />

40mg<br />

-6.2<br />

-3.2<br />

-12.7<br />

ATO<br />

80mg<br />

-6.4<br />

-3.4<br />

-12.2<br />

AML 10mg<br />

Difference versus<br />

Placebo (mmHg)<br />

Mean Change (mmHg)<br />

-9.8<br />

-16.2<br />

-10.7<br />

-15.9<br />

-12.3<br />

-16.1<br />

-9.7<br />

-16.3<br />

-9.2<br />

-17.6<br />

Difference versus<br />

Placebo (mmHg)<br />

-13.2<br />

-12.9<br />

-13.1<br />

-13.3<br />

-14.6<br />

Table 8. Efficacy of Combined Treatments in Reducing Diastolic Blood Pressure 1<br />

Parameter/Analysis<br />

ATO 0 mg<br />

(Placebo)<br />

ATO<br />

10mg<br />

ATO<br />

20mg<br />

AML 0mg Mean Change (mmHg) -3.3 -4.1 -3.9<br />

(Placebo)<br />

Difference versus<br />

- -0.8 -0.6<br />

Placebo (mmHg)<br />

AML 5mg Mean Change (mmHg) -7.6 -8.2 -9.4<br />

ATO<br />

40mg<br />

-5.1<br />

-1.8<br />

-7.3<br />

ATO<br />

80mg<br />

-4.1<br />

-0.8<br />

-8.4<br />

AML 10mg<br />

Difference versus<br />

Placebo (mmHg)<br />

Mean Change (mmHg)<br />

-4.3<br />

-10.4<br />

-4.9<br />

-9.1<br />

-6.1<br />

-10.6<br />

-4.0<br />

-9.8<br />

-5.1<br />

-11.1<br />

Difference versus<br />

Placebo (mmHg)<br />

-7.1<br />

-5.8<br />

-7.3<br />

-6.5<br />

-7.8<br />

Table 9. Efficacy of Combined Treatments in Reducing LDL-C (% Change from Baseline) 1<br />

Parameter/Analysis<br />

ATO 0mg<br />

(Placebo)<br />

ATO<br />

10mg<br />

ATO<br />

20mg<br />

ATO<br />

40mg<br />

ATO<br />

80mg<br />

AML 0mg Mean % Change -1.1 -33.4 -39.5 -43.1 -47.2<br />

(Placebo)<br />

AML 5mg Mean % Change -0.1 -38.7 -42.3 -44.9 -48.4<br />

AML 10mg Mean % Change -2.5 -36.6 -38.6 -43.2 -49.1<br />

370


Based on these results, the efficacy in terms of SBP and DBP reduction with<br />

atorvastatin/amlodipine versus therapy with amlodpine monotherapy was similar. In addition,<br />

with respect to LDL-C reduction, the efficacy with atorvastatin/amlodipine was similar to the<br />

efficacy of atorvastatin monotherapy.<br />

GEMINI Study 11<br />

Preliminary data of a randomized, open-label, noncomparative study evaluated the efficacy and<br />

safety of atorvastatin/amlodipine (Caduet ® ) as initial or added-on (integrated) therapy in the<br />

treatment of concomitant hyptertension and dyslipidemia (n=1,220). Subjects underwent lifestyle<br />

modifications and received atorvastatin/amlodipine (5/10, 10/10, 5/20, 10/20, 5/40, 10/40, 5/80, or<br />

10/80) daily with dose titration to improve blood pressure and lipid control. The primary endpoint<br />

was the percentage of subjects achieving goal blood pressure and LDL-C levels as established by<br />

the JNC VI and NCEP ATP III guidelines, respectively.<br />

All subjects (n=1,220) received study drug and had a mean blood pressure of 146.6±11.0/<br />

87.9±8.6mmHg and mean LDL-C level of 152.7±33.2mg/dL. At 14 weeks, 57.7% achieved both<br />

blood pressure and LDL-C goals. Therapy was well tolerated, with 4.8% discontinuing due to<br />

adverse events: 11.9% due to respiratory tract infections, 8.8% due to edema, 5.4% due to<br />

headache, and 4.3% due to myalgia.<br />

Additional Studies<br />

The AVALON study is a prospective, multi-center study which is evaluating the safety and<br />

efficacy of atorvastatin/amlodipine (Caduet ® ) versus placebo or either agent alone in the treatment<br />

of co-morbid hypertension and dyslipidemia (n=847). The only information available was<br />

presented at a Medical Conference. Data at eight weeks indicates significantly more subjects<br />

treated with atorvastatin/amlodipine attained goal LDL-C levels compared to subjects treated with<br />

amlodipine monotherapy (82.1% versus 12.4%), and significantly more subjects treated with<br />

atorvastatin/amlodipine attained goal BP compared to atorvastatin monotherapy (51.0% versus<br />

32.3%). In addition, significantly more patients treated with atorvastatin/amlodipine (45.6%)<br />

attained both goal LDL-C and BP, compared to those treated with amlodipine (8.3%) and<br />

atorvastatin (28.6%) monotherapies. This study did not compare atorvastatin/amlodipine<br />

(Caduet ® ) versus coadministration of amlodipine and atorvastatin monotherapies. (Note, written<br />

information provided by manufacturer based on Data on File).<br />

Additional Evidence<br />

Dose Simplification: No clinical data is available for the combination of<br />

atorvastatin/amlodipine (Caduet ® ), comparing adherence rates in patients given the<br />

combination agent versus therapy with each separate agent. The AVALON study<br />

presented above showed clinical benefit of the combination agent when compared with<br />

either agent as monotherapy, but no studies have compared the combination<br />

atorvastatin/amlodipine agent with use of coadministration of atorvastatin and<br />

amlodipine.<br />

Stable Therapy: A literature search of Medline and Ovid did not reveal clinical data on<br />

changing from other therapies to atorvastatin/amlodipine.<br />

Impact on Physician Visits: A literature search of Medline and Ovid did not reveal<br />

clinical data pertinent to use of atorvastatin/amlodipine and impact on physician visits.<br />

371


IX.<br />

Conclusions<br />

Recommended doses of atorvastatin/amlodipine have demonstrated dose-dependent reductions in<br />

both LDL-C and blood pressure which are both significant risk factors for the development of<br />

CHD. Efficacy and safety of this combination is similar to the individual agents administered<br />

separately (e.g., LDL-C reduction is similar to atorvastatin monotherapy, and BP reduction is<br />

similar to amlodipine monotherapy). No studies have compared the efficacy of<br />

atorvastatin/amlodipine to amlodipine and atorvastatin monotherapies administered<br />

concomitantly. Atorvastatin/amlodipine reduced LDL-C levels from ~33% to ~49% with the<br />

greatest reduction seen with the 80mg atorvastastin formulation. Systolic and diastolic BP were<br />

reduced by 12.2-17.6mmHg and 7.3-11.1mmHg. Preliminary data indicates, approximately 58%<br />

of subjects with co-morbid hyperlipidemia and hypertension achieved both BP and LDL-C goals<br />

based on current practice guidelines. Data regarding the efficacy of atorvastatin/amlodipine in the<br />

treatment of angina is not available in the Published Medical Literature. However, based on the<br />

similarity in pharmacokinetics of atorvastatin and amlodipine following atorvastatin/amlodipine<br />

(Caduet ® ) administration compared to atorvastatin and amlodipine pharmacokinetics when<br />

administered separately, theoretically, efficacy should be similar.<br />

One concern is with the available atorvastatin/amlodipine dosage forms. A formulation containing<br />

2.5mg of amlodipine is not available. This is the recommended starting dose for amlodipine in<br />

elderly and/or small, fragile individuals. This dose may also be used when adding amlodipine to<br />

other antihypertensive therapy. The combination product has no clinical advantage over<br />

amlodipine (Norvasc ® ) and atorvastatin (Lipitor ® ) in respect to BP reduction achieved with<br />

amlodipine (Norvasc ® ) and LDL-C lowering achieved with atorvastatin (Lipitor ® ).<br />

Therefore, atorvastatin/amlodipine (Caduet ® ) is comparable to the drugs in this class and to the<br />

generics and OTC products and offers no significant clinical advantage over other alternatives in<br />

general use.<br />

X. Recommendations<br />

No brand of the combination atorvastatin/amlodipine (Caduet ® ) is recommended for preferred<br />

status.<br />

372


References<br />

1. Caduet ® [package Insert]. NewYork, NY: Pfizer Labs. 2004.<br />

2. Grundy SM, Cleeman JI, Bairey Merz NC, et al for the Coordinating Committee of the<br />

National Cholesterol Education Program. NCEP report: implications of recent clinical<br />

trials for the National Cholesterol Education Program, Adult Treatment Panel III<br />

Guidelines. Circulation. 2004;110:227-239.<br />

3. Executive summary of the third report of the National Cholesterol Education Program<br />

(NCEP) Expert Panel on detection, evaluation, and treatment of high blood cholesterol in<br />

adults (adult treatment panel III). JAMA. 2001;285(19):2486-2497.<br />

4. US Department of Health and Human Services Food and Drug Administration Center for<br />

Drug Evaluation and Research Office of Pharmaceutical Science and Generic Drugs.<br />

Electronic Orange Book: Approved Drug Products with Therapeutic Equivalence<br />

Evaluations. Current through July 2004. Available at:<br />

http://www.fda.gov/cder/ob/default.htm. Accessed September 23, 2004.<br />

5. Chobanian AV, Bakris GL, Black HR, et al and the National High Blood Pressure<br />

Education Program Coordinating Committee. JNC 7: seventh report of the Joint National<br />

Committee on prevention, detection, evaluation, and treatment of high blood pressure.<br />

Hypertension. 2003;42:1206-1252.<br />

6. US Department of Health and Human Services. Seventh Report of the Joint National<br />

Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure.<br />

Available at: http://www.nhlbi.nih.gov/guidelines/hypertension/express.pdf. Accessed<br />

September 23, 2004.<br />

7. Gibbons RJ, Abrams J, Chatterjee K, et al. ACC/AHA 2002 guideline update for the<br />

management of patients with chronic stable angina: a report of the American College of<br />

Cardiology/American Heart Association Task Force on Practice Guidelines (Committee<br />

to Update the 1999 Guidelines for the Management of Patients with Chronic Stable<br />

Angina). 2002. Available at www.acc.org/clinical/guidelines/stable/stable.pdf.<br />

8. Lipitor ® [package insert]. New York, NY: Pfizer Labs. July 2004.<br />

9. Norvasc ® [package insert]. New York, NY: Pfizer Labs. June 2003.<br />

10. Facts and Comparisons Online. 2004. Wolters Kluwer Health, Inc. Available at:<br />

http://www.efactsweb.com/index.asp. Accessed September 23, 2004.<br />

11. Blank R, LaSalle J, Reeves R, Piper BA, Sun F. Amlodipine/atorvastatin single pill dual<br />

therapy improves goal attainment in the treatment of concomitant hypertension and<br />

dyslipidemia: the GEMINI study [abstract]. J Am Coll Cardiol. 2004;43(suppl A):447A.<br />

373

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!