22.05.2022 Views

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

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

less potent and substantially more toxic than the pure (−)-enantiomer.

Compared with the (+)-enantiomer, the phosphorylated (−)-

enantiomer is more resistant to cleavage from nascent RNA/DNA

duplexes by cellular 3′-5′ exonucleases, which may contribute to its

greater potency. The IC 50

of lamivudine against laboratory strains of

HIV-1 ranges from 2 to 670 nM, although the IC 50

in primary human

peripheral blood mononuclear cells is as high as 15 μM (Perry and

Faulds, 1997).

Mechanisms of Action and Resistance. Lamivudine

enters cells by passive diffusion, is converted to the

monophosphate by deoxycytidine kinase, and undergoes

further phosphorylation by deoxycytidine monophosphate

kinase and nucleoside diphosphate kinase to yield

lamivudine 5′-triphosphate, which is the active anabolite

(Perry and Faulds, 1997) (Figure 59–3). Lamivudine is

phosphorylated more efficiently in resting cells, which

may explain its reduced potency in primary peripheral

blood mononuclear cells as compared with cell lines

(Gao et al., 1994). Lamivudine has low affinity for

human DNA polymerases, explaining its low toxicity to

the host.

High-level resistance to lamivudine occurs with single-aminoacid

substitutions, M184V or M184I. These mutations can reduce in

vitro sensitivity to lamivudine by up to 1000-fold (Perry and Faulds,

1997). The same mutations confer high-level cross-resistance to

emtricitabine and a lesser degree of resistance to abacavir (Gallant et

al., 2003). The M184V mutation restores zidovudine susceptibility in

zidovudine-resistant HIV (Larder et al., 1995) and also partially

restores tenofovir susceptibility in tenofovir-resistant HIV harboring

the K65R mutation (Wainberg et al., 1999). The same K65R mutation

confers resistance to lamivudine, emtricitabine, didanosine, stavudine,

and abacavir.

HIV-1 isolates harboring the M184V mutation have increased

transcriptional fidelity in vitro (Wainberg et al., 1996) and decreased

replication capacity (Miller et al., 1999). Variants with the M184I

mutation are even more impaired with regard to in vitro replication

(Larder et al., 1995) and usually are replaced in lamivudine-treated

patients by the M184V mutation. The reduced fitness of lamivudineresistant

viruses harboring these mutations, and their ability to prevent

or partially reverse the effect of thymidine analog mutations,

may contribute to the sustained virologic benefits of zidovudine and

lamivudine combination therapy (Eron et al., 1995).

Lamivudine is used to treat HBV infection (Chapter 58), and

some parallels in drug resistance are worth noting. High-level resistance

to lamivudine occurs with a single mutation in the HBV DNA

polymerase gene; as with HIV, this consists of a methionine-tovaline

substitution (M2041V) in the enzyme active site. Resistance

to lamivudine occurs in up to 90% of HIV/HBV co-infected patients

after 4 years of treatment. However, virologic benefits persist in

some treated patients harboring lamivudine-resistant HBV possibly

because the mutated virus has substantially reduced replicative

capacity (Leung et al., 2001).

Absorption, Distribution, and Elimination. The oral bioavailability

of lamivudine is >80% and is not affected by food. Although

lamivudine was marketed originally with a recommended dose of

150 mg twice daily based on the short plasma t 1/2

of the parent compound,

the intracellular t 1/2

of lamivudine 5′-triphosphate is 12-18

hours, and the drug is now approved for use once daily at 300 mg

(Moore et al., 1999).

Lamivudine is excreted primarily unchanged in the urine, and

dose adjustment is recommended for patients with a creatinine clearance

<50 mL/minute (Jayasekara et al., 1999). Lamivudine does not

bind significantly to plasma proteins and freely crosses the placenta

into the fetal circulation. Like zidovudine, lamivudine concentrations

are higher in the male genital tract than in the peripheral circulation,

suggesting active transport or trapping. Penetration to the

CNS appears to be moderate, with a CSF-to-plasma concentration

ratio of ≤0.15 (Perry and Faulds, 1997). The clinical significance of

the low CSF penetration is unknown.

Untoward Effects. Lamivudine is one of the least toxic antiretroviral

drugs and has few significant adverse effects.

Neutropenia, headache, and nausea have been reported at higher

than recommended doses. Pancreatitis has been reported in pediatric

patients, but this has not been confirmed in controlled trials of adults or

children. Because lamivudine also has activity against HBV and substantially

lowers plasma HBV DNA concentrations, caution is warranted

in using this drug in patients co-infected with HBV or in

HBV-endemic areas; discontinuation of lamivudine may be associated

with a rebound of HBV replication and exacerbation of hepatitis.

Precautions and Interactions. Because lamivudine and emtricitabine

have nearly identical resistance and activity patterns, there is no

rationale for their combined use. Lamivudine is synergistic with

most other nucleoside analogs in vitro (Perry and Faulds, 1997).

Therapeutic Use. Lamivudine (EPIVIR) is approved for

HIV in adults and children ≥3 months of age.

In early monotherapy studies, initial declines in plasma HIV-1

RNA concentrations of up to 90% occurred within 14 days but

rebounded rapidly with emergence of lamivudine-resistant HIV

(Perry and Faulds, 1997). Patients randomized to the combination

of lamivudine plus zidovudine had substantially better mean

decreases in plasma HIV-1 RNA at 52 weeks (97% vs. 70% decrease

in copies/mL) and increases in CD4+ lymphocyte counts (+61 versus

–53 cells/mm 3 ) compared with those receiving zidovudine alone

(Eron et al., 1995). In a large randomized double-blind trial, combining

lamivudine with zidovudine or stavudine caused ~12-fold further

decline in viral load at 24 weeks compared with zidovudine or

stavudine monotherapy (Kuritzkes et al., 1999); in the same trial,

combining lamivudine with didanosine conferred no additional benefits.

Lamivudine has been effective in combination with other antiretroviral

drugs in both treatment-naive and experienced patients

(Perry and Faulds, 1997) and is a common component of therapy,

given its safety, convenience, and efficacy.

Lamivudine (EPIVIR-HBV) also is approved for treatment of

chronic hepatitis B.

Abacavir

Chemistry and Antiviral Activity. Abacavir is a synthetic

carbocyclic purine analog (Figure 59–2).

1635

CHAPTER 59

ANTIRETROVIRAL AGENTS AND TREATMENT OF HIV INFECTION

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

Saved successfully!

Ooh no, something went wrong!