22.05.2022 Views

DƯỢC LÍ Goodman & Gilman's The Pharmacological Basis of Therapeutics 12th, 2010

Create successful ePaper yourself

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

1636 Carbovir, a related guanine analog, was withdrawn from clinical

development owing to poor oral bioavailability (Hervey and

Perry, 2000). The IC 50

of abacavir for primary clinical HIV-1 isolates

is 0.26 μM, and its IC 50

for laboratory strains ranges from 0.07

to 5.8 μM.

SECTION VII

CHEMOTHERAPY OF MICROBIAL DISEASES

Mechanisms of Action and Resistance. Abacavir is the

only approved antiretroviral that is active as a guanosine

analog. It is initially monophosphorylated by

adenosine phosphotransferase. The monophosphate is

then converted to (−)-carbovir 3′-monophosphate,

which is then phosphorylated to the di- and triphosphates

by cellular kinases (Figure 59–3). Carbovir

5′-triphosphate terminates the elongation of proviral

DNA because it is incorporated by reverse transcriptase

into nascent DNA but lacks a 3′-hydroxyl group.

Clinical resistance to abacavir is associated with four specific

codon substitutions: K65R, L74V, Y115F, and M184V (Gallant et al.,

2003). Individually, these substitutions produce only modest (2- to

4-fold) resistance to abacavir, but in combination can reduce susceptibility

by up to 10-fold. The Y115F mutation is seen uniquely with

abacavir and causes low-level resistance. The L74V mutation is associated

with cross-resistance to the purine analog didanosine. K65R

confers cross-resistance to all nucleosides except zidovudine. An

alternate pathway for abacavir resistance involves mutations at

codons 41, 210, and 215, which have been associated with a

reduced likelihood of virologic response. Abacavir sensitivity is

greatly reduced by the multinucleoside resistance clusters, including

that associated with the Q151M, as well as the 2-amino acid

insertion following codon 69 (Gallant et al., 2003).

Absorption, Distribution, and Elimination. Abacavir’s oral

bioavailability is >80% regardless of food intake (Table 59–2).

Abacavir is eliminated by metabolism to the 5′-carboxylic acid

derivative catalyzed by alcohol dehydrogenase, and by glucuronidation

to the 5′-glucuronide (Figure 59–3). These metabolites account

for 30% and 36% of elimination, respectively (Hervey and Perry,

2000). Abacavir is not a substrate or inhibitor of CYPs. Abacavir is

50% bound to plasma proteins, and the CSF/plasma AUC ratio is

~0.3. Although the introduced dose of abacavir was 300 mg twice

daily, carbovir triphosphate accumulates inside the cell and has a

reported elimination t 1/2

of up to 21 hours (Hervey and Perry, 2000);

thus a regimen of 600 mg once daily is approved now.

Untoward Effects. The most important adverse effect of abacavir is

a unique and potentially fatal hypersensitivity syndrome. This syndrome

is characterized by fever, abdominal pain, and other gastrointestinal

(GI) complaints; a mild maculopapular rash; and malaise or

fatigue. Respiratory complaints (cough, pharyngitis, dyspnea), musculoskeletal

complaints, headache, and paresthesias are reported less

commonly. Median time to onset of symptoms is 11 days, and 93% of

cases occur within 6 weeks of initiating therapy (Hetherington et al.,

2002). The presence of concurrent fever, abdominal pain, and rash

within 6 weeks of starting abacavir is diagnostic and necessitates

immediate discontinuation of the drug. Patients having only one of

these symptoms may be observed to see if additional symptoms

appear. Unlike many hypersensitivity syndromes, this condition

worsens with continued treatment. Abacavir can never be restarted

once discontinued for hypersensitivity because reintroduction of the

drug leads to rapid recurrence of severe symptoms, accompanied by

hypotension, a shocklike state, and possibly death. The reported mortality

rate of restarting abacavir in sensitive individuals is 4% (Hervey

and Perry, 2000).

Abacavir hypersensitivity occurs in 2-9% of patients depending

on the population studied. The cause is a genetically mediated

immune response linked to both the HLA-B*5701 locus and the

M493T allele in the heat-shock locus Hsp70-Hom (Mallal et al.,

2008). The latter gene is implicated in antigen presentation, and this

haplotype is associated with aberrant tumor necrosis factor-α

release after exposure of human lymphocytes to abacavir ex vivo.

This is one of the strongest pharmacogenetic associations ever

described. In one white population, the combination of these two

markers occurred in 94.4% of cases and <0.5% of controls for a

positive predictive value of 93.8% and a negative predictive value

of 99.5% (Mallal et al., 2008). Abacavir should not be given to those

with the HLA-B*5701 genotype; in all others, the risk of true

hypersensitivity is essentially zero (Mallal et al., 2008). Aside

from hypersensitivity, abacavir is a well-tolerated drug. Carbovir

5′-triphosphate is a weak inhibitor of human DNA polymerases,

including DNA polymerase-γ (Hervey and Perry, 2000). Abacavir

therefore has not been associated with adverse events thought to be

due to mitochondrial toxicity. Epidemiological associations link

abacavir use and increased risk of myocardial infarction (D:A:D

Study Group et al., 2008).

Precautions and Interactions. Abacavir is not associated with any

clinically significant pharmacokinetic drug interactions. However,

a large dose of ethanol (0.7 g/kg) increased the abacavir plasma

AUC by 41% and prolonged the elimination t 1/2

by 26%

(McDowell et al., 2000) possibly owing to competition for alcohol

dehydrogenase.

Therapeutic Use. Abacavir (ZIAGEN) is approved for the

treatment of HIV-1 infection, in combination with other

antiretroviral agents. In initial monotherapy studies,

abacavir reduced HIV plasma RNA concentrations up

to 300 times more than that seen with other antiretroviral

nucleosides, and it increased CD4+ lymphocyte

counts by 80-200 cells/mm 3 (Hervey and Perry, 2000).

Abacavir is not a more potent inhibitor of HIV replication

than other nucleosides in vitro, and the mechanism

for its more potent in vivo monotherapy activity is

unexplained.

Abacavir is effective in combination with other nucleoside

analogs, NNRTIs, and protease inhibitors. Adding abacavir to

zidovudine and lamivudine resulted in a substantially greater

decrease in plasma HIV-l RNA than seen with the two-drug regimen

of zidovudine plus lamivudine in adults or children (Hervey and

Perry, 2000). Abacavir is available in a co-formulation with zidovudine

and lamivudine (TRIZIVIR) for twice-daily dosing. However, the

combination of abacavir, zidovudine, and lamivudine was less effective

in a randomized, double-blind, placebo-controlled trial in treatment-naive

patients than was the combination of zidovudine,

lamivudine, and efavirenz or the four-drug regimen of zidovudine,

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!