You are on page 1of 34

Antiviral Agents

Viruses are obligate intracellular parasites; their


replication depends primarily on synthetic
processes of the host cell.
Therefore, to be effective, antiviral agents must
either block viral entry into or exit from the cell
or be active inside the host cell.
As a corollary, nonselective inhibitors of virus
replication may interfere with host cell function
and result in toxicity.
Progress in antiviral chemotherapy began in the
early 1950s, when the search for anti-cancer
drugs generated several new compounds capable
of inhibiting viral DNA synthesis.
The two first-generation antiviral agents, 5-
iododeoxyuridine and trifluorothymidine, had
poor specificity (ie, they inhibited host cell DNA
as well as viral DNA) that rendered them too
toxic for systemic use.
However, both agents are effective when used
topically for the treatment of herpes keratitis.
Knowledge of the mechanisms of viral
replication has provided insights into critical
steps in the viral life cycle that can serve as
Viral replication requires several steps (Figure
potential targets for antiviral therapy.
49–1):
Recent research has focused on identifying
(1) attachment of the virus to receptors on the
agents with greater selectivity, higher potency,
host cell surface;
in vivo stability, and reduced toxicity.
(2) entry of the virus through the host cell
Antiviral therapy is now available for
membrane;
herpesviruses, hepatitis C virus (HCV), hepatitis
(3) uncoating of viral nucleic acid;
B virus (HBV), papillomavirus, influenza,
(4) synthesis of early regulatory proteins, eg,
human immunodeficiency virus (HIV), and
nucleic acid polymerases;
respiratory syncytial virus (RSV).
(5) synthesis of new viral RNA or DNA;
Antiviral drugs share the common property of
(6) integration into the nuclear genome;
being virustatic; they are active only against
(7) synthesis of late, structural proteins;
replicating viruses and do not affect latent virus.
(8) assembly (maturation) of viral particles; and
Whereas some infections require monotherapy
(9) release from the cell.
for brief periods of time (eg, acyclovir for
Antiviral agents can potentially target any of
herpes simplex virus), others require dual
these steps.
therapy for prolonged periods of time (interferon
alfa/ribavirin for HCV), whereas still others
require multiple drug therapy for indefinite
periods (HIV).
In chronic illnesses such as viral hepatitis and
HIV infection, potent inhibition of viral
replication is crucial in limiting the extent of
systemic damage.
and valacyclovir for the treatment of herpes
zoster infections.

ACYCLOVIR
Acyclovir (eFigure 49–1.1) is an acyclic
guanosine derivative with clinical activity
against HSV-1, HSV-2, and VZV, but it is
approximately 10 times more potent against
HSV-1 and HSV-2 than against VZV.
In vitro activity against Epstein-Barr virus
(EBV), cytomegalovirus (CMV), and human
herpesvirus-6 (HHV-6) is present but weaker.
Acyclovir requires three phosphorylation
steps for activation.
It is converted first to the monophosphate
derivative by the virus-specified thymidine
kinase and then to the di- and triphosphate
compounds by host cell enzymes (Figure 49–
2).
Because it requires the viral kinase for initial
phosphorylation, acyclovir is selectively
activated—and the active metabolite
accumulates—only in infected cells.
Acyclovir triphosphate inhibits viral DNA
synthesis by two mechanisms: competition
with deoxyGTP for the viral DNA
polymerase, resulting in binding to the DNA
template as an irreversible complex; and chain
termination following incorporation into the
viral DNA.
The bioavailability of oral acyclovir is low
(15–20%) and is unaffected by food.
An intravenous formulation is available.
Topical formulations produce high
concentrations in herpetic lesions, but
systemic concentrations are undetectable by
this route.
Acyclovir is cleared primarily by glomerular
filtration and tubular secretion.
AGENTS TO TREAT HERPES SIMPLEX VIRUS
The half-life is 2.5–3 hours in patients with
(HSV) & VARICELLA-ZOSTER VIRUS (VZV)
normal renal function and 20 hours in patients
INFECTIONS with anuria.
Acyclovir diffuses readily into most tissues
Three oral nucleoside analogs are licensed for and body fluids.
the treatment of HSV and VZV infections: Cerebrospinal fluid concentrations are 20–
acyclovir, valacyclovir, and famciclovir. 50% of serum values.
They have similar mechanisms of action and Oral acyclovir has multiple uses. In first
comparable indications for clinical use; all are episodes of genital herpes, oral acyclovir
well tolerated. shortens the duration of symptoms by
Acyclovir has been the most extensively studied; approximately 2 days, the time to lesion
it was licensed first and is the only one of the healing by 4 days, and the duration of viral
shedding by 7 days.
three that is available for intravenous use in the
In recurrent anogenital herpes, the time course
United States. is shortened by 1–2 days.
Comparative trials have demonstrated similar Treatment of first-episode genital herpes does
efficacies of these three agents for the treatment not alter the frequency or severity of recurrent
of HSV but modest superiority of famciclovir outbreaks.
Long-term suppression with oral acyclovir in famciclovir, and ganciclovir.
patients with frequent recurrences of genital Agents such as foscarnet, cidofovir, and
herpes decreases the frequency of trifluridine do not require activation by viral
symptomatic recurrences and of thymidine kinase and thus have preserved
asymptomatic viral shedding, thus decreasing activity against the most prevalent acyclovir-
the rate of sexual transmission. resistant strains (Figure 49–2).
However, outbreaks may resume upon Acyclovir is generally well tolerated, although
discontinuation of suppressive acyclovir. nausea, diarrhea, and headache may occur.
Oral acyclovir is only modestly beneficial in Intravenous infusion may be associated with
recurrent herpes labialis. reversible renal toxicity (ie, crystalline
In contrast, acyclovir therapy significantly nephropathy or interstitial nephritis) or
decreases the total number of lesions, duration neurologic effects (eg, tremors, delirium,
of symptoms, and viral shedding in patients seizures).
with varicella (if begun within 24 hours after However, these are uncommon with adequate
the onset of rash) or cutaneous zoster (if hydration and avoidance of rapid infusion
begun within 72 hours); the risk of post- rates.
herpetic neuralgia is also reduced if treatment High doses of acyclovir cause chromosomal
is initiated early. damage and testicular atrophy in rats, but
However, because VZV is less susceptible to there has been no evidence of teratogenicity,
acyclovir than HSV, higher doses are required reduction in sperm production, or cytogenetic
(Table 49–1). alterations in peripheral blood lymphocytes in
When given prophylactically to patients patients receiving daily suppression of genital
undergoing organ transplantation, oral or herpes for more than 10 years.
intravenous acyclovir prevents reactivation of A recent study found no evidence of increased
HSV and VZV infection. birth defects in 1150 infants who were
Evidence from clinical trials suggests that the exposed to acyclovir during the first trimester.
use of daily acyclovir (400 mg twice daily) In fact, the American College of Obstetricians
may reduce the plasma viral load of HIV-1 and Gynecologists recommends suppressive
and the risk of HIV-associated disease acyclovir therapy beginning at week 36 in
progression in individuals dually infected pregnant women with active recurrent genital
with HSV-2 and HIV-1. herpes to reduce the risk of recurrence at
Intravenous acyclovir is the treatment of delivery and possibly the need for cesarean
choice for herpes simplex encephalitis, section.
neonatal HSV infection, and serious HSV or The impact of this intervention on neonatal
VZV infections (Table 49–1). infection has not been established.
In neonates with central nervous system HSV, Concurrent use of nephrotoxic agents may
oral acyclovir suppression for 6 months enhance the potential for nephrotoxicity.
following acute treatment improves Probenecid and cimetidine decrease acyclovir
neurodevelopmental outcomes. clearance and increase exposure.
In immunocompromised patients with VZV Somnolence and lethargy may occur in
infection, intravenous acyclovir reduces the patients receiving concomitant zidovudine
incidence of cutaneous and visceral and acyclovir.
dissemination.
Topical acyclovir cream is substantially less
effective than oral therapy for primary HSV
infection.
It is of no benefit in treating recurrent genital
herpes.
Resistance to acyclovir can develop in HSV
or VZV through alteration in either the viral
thymidine kinase or the DNA polymerase,
and clinically resistant infections have been
reported in immunocompromised hosts.
Most clinical isolates are resistant on the basis
of deficient thymidine kinase activity and thus
are cross-resistant to valacyclovir,
with intravenous acyclovir.
Oral bioavailability is 54–70%, and
cerebrospinal fluid levels are about 50% of
those in serum.
Elimination half-life is 2.5–3.3 hours.
Twice-daily valacyclovir is effective for
treatment of first or recurrent genital herpes
and varicella and zoster infections; it is
approved for use as a 1-day treatment for
orolabial herpes and as suppression of
frequently recurring genital herpes (Table 49–
1).
Once-daily dosing of valacyclovir for chronic
suppression in persons with recurrent genital
herpes has been shown to markedly decrease
the risk of sexual transmission.
In comparative trials with acyclovir for the
treatment of patients with zoster, rates of
cutaneous healing were similar, but
valacyclovir was associated with a shorter
duration of zoster-associated pain.
Higher doses of valacyclovir (2 g four times
daily) are effective in preventing CMV
disease after organ transplantation and
suppressive valacyclovir prevents VZV
reactivation after hematopoietic stem cell
transplantation.
Valacyclovir is generally well tolerated,
although nausea, headache, vomiting, or rash
may occur.
At high doses, confusion, hallucinations, and
seizures have been reported.
AIDS patients who received high-dosage
valacyclovir chronically (ie, 8 g/d) had
increased gastrointestinal intolerance as well
as thrombotic thrombocytopenic
purpura/hemolytic-uremic syndrome; this
dose has also been associated with confusion
and hallucinations in transplant patients. In a
recent study, there was no evidence of
increased birth defects in 181 infants who
were exposed to valacyclovir during the first
trimester.

FAMCICLOVIR
VALACYCLOVIR Famciclovir is the diacetyl ester prodrug of 6-
Valacyclovir is the L-valyl ester of acyclovir. deoxypenciclovir, an acyclic guanosine
It is rapidly converted to acyclovir after oral analog (eFigure 49–2.1).
administration via first-pass enzymatic After oral administration, famciclovir is
hydrolysis in the liver and intestine, resulting rapidly deacetylated and oxidized by first-
in serum levels that are three to five times pass metabolism to penciclovir.
greater than those achieved with oral It is active in vitro against HSV-1, HSV-2,
acyclovir and approximate those achieved VZV, EBV, and HBV.
As with acyclovir, activation by
phosphorylation is catalyzed by the virus- duration of recurrent herpes labialis by ~ 17
specified thymidine kinase in infected cells, hours compared to placebo when applied
followed by competitive inhibition of the viral within 1 hour of the onset of prodromal
DNA polymerase to block DNA synthesis. symptoms and continued every 2 hours during
Unlike acyclovir, however, penciclovir does waking hours for 4 days.
not cause chain termination. Adverse effects are uncommon, other than
Penciclovir triphosphate has lower affinity for application site reactions in ~1%.
the viral DNA polymerase than acyclovir
triphosphate, but it achieves higher
intracellular concentrations. DOCOSANOL
The most commonly encountered clinical Docosanol is a saturated 22-carbon aliphatic
mutants of HSV are thymidine kinase- alcohol that inhibits fusion between the host
deficient; these are cross-resistant to acyclovir cell plasma membrane and the HSV envelope,
and famciclovir. thereby preventing viral entry into cells and
The bioavailability of penciclovir from orally subsequent viral replication.
administered famciclovir is 70%. Topical docosanol 10% cream is available
The intracellular half-life of penciclovir without a prescription.
triphosphate is prolonged, at 7–20 hours. When applied within 12 hours of the onset of
Penciclovir is excreted primarily in the urine. prodromal symptoms, five times daily,
Oral famciclovir is effective for the treatment median healing time was shortened by 18
of first and recurrent genital herpes, for hours compared with placebo in recurrent
chronic daily suppression of genital herpes, orolabial herpes.
for treatment of herpes labialis, and for the Application site reactions occur in ~2%.
treatment of acute zoster (Table 49–1).
One-day usage of famciclovir significantly
accelerates time to healing of recurrent genital TRIFLURIDINE
herpes and of herpes labialis. Trifluridine (trifluorothymidine) is a
Comparison of famciclovir to valacyclovir for fluorinated pyrimidine nucleoside that inhibits
treatment of herpes zoster in viral DNA synthesis in HSV-1, HSV-2,
immunocompetent patients showed similar CMV, vaccinia, and some adenoviruses.
rates of cutaneous healing and pain It is phosphorylated intracellularly by host
resolution; both agents shortened the duration cell enzymes, and then competes with
of zoster-associated pain compared with thymidine triphosphate for incorporation by
acyclovir. the viral DNA polymerase (Figure 49–2).
Oral famciclovir is generally well tolerated, Incorporation of trifluridine triphosphate into
although headache, nausea, or diarrhea may both viral and host DNA prevents its systemic
occur. use.
As with acyclovir, testicular toxicity has been Application of a 1% solution is effective in
demonstrated in animals receiving repeated treating keratoconjunctivitis and recurrent
doses. epithelial keratitis due to HSV-1 or HSV-2.
However, men receiving daily famciclovir Cutaneous application of trifluridine solution,
(250 mg every 12 hours) for 18 weeks had no alone or in combination with interferon alfa,
changes in sperm morphology or motility. has been used successfully in the treatment of
In a recent study, there was no evidence of acyclovir-resistant HSV infections.
increased birth defects in 32 infants who were
exposed to famciclovir during the first INVESTIGATIONAL AGENTS
trimester. Valomaciclovir is an inhibitor of the viral
The incidence of mammary adenocarcinoma DNA polymerase; it is currently under
was increased in female rats receiving clinical evaluation for the treatment of
famciclovir for 2 years. patients with acute zoster and acute EBV
infection (infectious mononucleosis).
PENCICLOVIR
The guanosine analog penciclovir, the active AGENTS TO TREAT CYTOMEGALOVIRUS
metabolite of famciclovir, is available for (CMV) INFECTIONS
topical use.
Penciclovir cream (1%) shortened the median
CMV infections occur primarily in the setting of creatinine clearance.
advanced immunosuppression and are typically Ganciclovir is readily cleared by
due to reactivation of latent infection. hemodialysis.
Dissemination of infection results in end-organ Intravenous ganciclovir has been shown to
disease, including retinitis, colitis, esophagitis, delay progression of CMV retinitis in
central nervous system disease, and immunocompromised patients.
Dual therapy with foscarnet and ganciclovir is
pneumonitis.
more effective in delaying progression of
Although the incidence in HIV-infected patients
retinitis than either drug alone in patients with
has markedly decreased with the advent of AIDS (see Foscarnet), although adverse
potent anti-retroviral therapy, clinical effects are compounded.
reactivation of CMV infection after organ Intravenous ganciclovir is also used to treat
transplantation is still prevalent. CMV colitis, esophagitis, and pneumonitis
The availability of oral valganciclovir has (the latter often in combination with
decreased the use of intravenous ganciclovir, intravenous cytomegalovirus
intravenous foscarnet, and intravenous cidofovir immunoglobulin) in immunocompromised
for the prophylaxis and treatment of end-organ patients.
CMV disease (Table 49–2). Intravenous ganciclovir, followed by either
Oral valganciclovir has replaced oral oral ganciclovir or high-dose oral acyclovir,
ganciclovir because of its lower pill burden. reduced the risk of CMV infection in
transplant recipients.
Limited data in infants with symptomatic
congenital neurologic CMV disease suggest
that treatment with IV ganciclovir may reduce
hearing loss.
The risk of Kaposi’s sarcoma is reduced in
AIDS patients receiving long-term
ganciclovir, presumably because of activity
against HHV-8.
GANCICLOVIR Intravitreal injections of ganciclovir may be
Ganciclovir is an acyclic guanosine analog used to treat CMV retinitis.
(eFigure 49–2.1) that requires activation by Concurrent therapy with a systemic anti-CMV
triphosphorylation before inhibiting the viral agent is necessary to prevent other sites of
DNA polymerase. end-organ CMV disease.
Initial phosphorylation is catalyzed by the The intraocular ganciclovir implant is no
virus-specified protein kinase longer available in the USA. Resistance to
phosphotransferase UL97 in CMV-infected ganciclovir increases with duration of use.
cells. The more common mutation, in UL97, results
The activated compound competitively in decreased levels of the triphosphorylated
inhibits viral DNA polymerase and causes (ie, active) form of ganciclovir.
termination of viral DNA elongation (Figure The less common UL54 mutation in DNA
49–2). polymerase results in higher levels of
Ganciclovir has in vitro activity against CMV, resistance and potential cross-resistance with
HSV, VZV, EBV, HHV-6, and HHV-8. Its cidofovir and foscarnet.
activity against CMV is up to 100 times Antiviral susceptibility testing is
greater than that of acyclovir. recommended in patients in whom resistance
Ganciclovir is administered intravenously; the is suspected clinically.
bioavailability of oral ganciclovir is poor, and The most common adverse effect of
it is no longer available in the US. intravenous ganciclovir treatment is
Ganciclovir gel is available for the treatment myelosuppression, which although reversible
of acute herpetic keratitis. may be dose-limiting.
Cerebrospinal fluid concentrations are Myelosuppression may be additive in patients
approximately 50% of serum concentrations. receiving concurrent zidovudine,
The elimination half-life is 4 hours, and the azathioprine, or mycophenolate mofetil.
intracellular half-life is prolonged at 16–24 Other potential adverse effects are nausea,
hours. diarrhea, fever, rash, headache, insomnia, and
Clearance of the drug is linearly related to
peripheral neuropathy. Foscarnet blocks the pyrophosphate binding
Central nervous system toxicity (confusion, site of these enzymes and inhibits cleavage of
seizures, psychiatric disturbance) and pyrophosphate from deoxynucleotide
hepatotoxicity have been rarely reported. triphosphates.
Intravitreal ganciclovir has been associated It has in vitro activity against HSV, VZV,
with vitreous hemorrhage and retinal CMV, EBV, HHV-6, HHV-8, HIV-1, and
detachment. HIV-2.
Ganciclovir is mutagenic in mammalian cells Foscarnet is available in an intravenous
and carcinogenic and embryotoxic at high formulation only; poor oral bioavailability
doses in animals and causes and gastrointestinal intolerance preclude oral
aspermatogenesis; the clinical significance of use.
these preclinical data is unclear. Cerebrospinal fluid concentrations are 43–
Levels of ganciclovir may rise in patients 67% of steady-state serum concentrations.
concurrently taking probenecid or Although the mean plasma half-life is 3–7
trimethoprim. hours, up to 30% of foscarnet may be
Concurrent use of ganciclovir with didanosine deposited in bone, with a half-life of several
may result in increased levels of didanosine. months.
The clinical repercussions of this are
unknown.
VALGANCICLOVIR Clearance of foscarnet is primarily renal and
Valganciclovir is an L-valyl ester prodrug of is directly proportional to creatinine
ganciclovir that exists as a mixture of two clearance.
diastereomers. Serum drug concentrations are reduced
After oral administration, both diastereomers approximately 50% by hemodialysis.
are rapidly hydrolyzed to ganciclovir by Foscarnet is effective in the treatment of end-
esterases in the intestinal wall and liver. organ CMV disease (ie, retinitis, colitis, and
Valganciclovir is well absorbed; the esophagitis), including ganciclovir-resistant
bioavailability of oral valganciclovir is 60% disease; it is also effective against acyclovir-
and it is recommended that the drug be taken resistant HSV and VZV infections.
with food. The dosage of foscarnet must be titrated
The AUC0–24h resulting from valganciclovir according to the patient’s calculated
(900 mg once daily) is similar to that after 5 creatinine clearance before each infusion.
mg/kg once daily of intravenous ganciclovir Use of an infusion pump to control the rate of
and approximately 1.65 times that of oral infusion is important to prevent toxicity, and
ganciclovir. large volumes of fluid are required because of
The major route of elimination is renal, the drug’s poor solubility.
through glomerular filtration and active The combination of ganciclovir and foscarnet
tubular secretion. is synergistic in vitro against CMV and has
Plasma concentrations of valganciclovir are been shown to be superior to either agent
reduced approximately 50% by hemodialysis. alone in delaying progression of retinitis;
Valganciclovir is as effective as intravenous however, toxicity is also increased when these
ganciclovir for the treatment of CMV retinitis agents are administered concurrently.
and is also indicated for the prevention of As with ganciclovir, a decrease in the
CMV disease in high-risk solid organ and incidence of Kaposi’s sarcoma has been
bone marrow transplant recipients. observed in patients who have received long-
Adverse effects, drug interactions, and term foscarnet.
resistance patterns are the same as those Foscarnet has been administered intravitreally
associated with ganciclovir. for the treatment of CMV retinitis in patients
with AIDS, but data regarding efficacy and
safety are incomplete.
FOSCARNET Resistance to foscarnet in HSV and CMV
Foscarnet (phosphonoformic acid) is an isolates is due to point mutations in the DNA
inorganic pyrophosphate analog that inhibits polymerase gene and is typically associated
herpesvirus DNA polymerase, RNA with prolonged or repeated exposure to the
polymerase, and HIV reverse transcriptase drug.
directly without requiring activation by Mutations in the HIV-1 reverse transcriptase
phosphorylation.
gene have also been described. allowing infrequent dosing.
Although foscarnet-resistant CMV isolates A separate metabolite, cidofovir
are typically cross-resistant to ganciclovir, phosphocholine, has a half-life of at least 87
foscarnet activity is usually maintained hours and may serve as an intracellular
against ganciclovir- and cidofovir-resistant reservoir of active drug.
isolates of CMV. Cerebrospinal fluid penetration is poor.
Potential adverse effects of foscarnet include Elimination is by active renal tubular
renal impairment, hypo- or hypercalcemia, secretion.
hypo- or hyperphosphatemia, hypokalemia, High-flux hemodialysis reduces serum levels
and hypomagnesemia. of cidofovir by approximately 75%.
Saline preloading helps prevent Intravenous cidofovir is effective for the
nephrotoxicity, as does avoidance of treatment of CMV retinitis and is used
concomitant administration of drugs with experimentally to treat adenovirus, human
nephrotoxic potential (eg, amphotericin B, papillomavirus, BK polyomavirus, vaccinia,
pentamidine, aminoglycosides). and poxvirus infections.
The risk of severe hypocalcemia, caused by Intravenous cidofovir must be administered
chelation of divalent cations, is increased with with high-dose probenecid (2 g at 3 hours
concomitant use of pentamidine. before the infusion and 1 g at 2 and 8 hours
Genital ulcerations associated with foscarnet after), which blocks active tubular secretion
therapy may be due to high levels of ionized and decreases nephrotoxicity.
drug in the urine. Before each infusion, cidofovir dosage must
Nausea, vomiting, anemia, elevation of liver be adjusted for alterations in the calculated
enzymes, and fatigue have been reported; the creatinine clearance or for the presence of
risk of anemia may be additive in patients urine protein, and aggressive adjunctive
receiving concurrent zidovudine. hydration is required.
Central nervous system toxicity includes Initiation of cidofovir therapy is
headache, hallucinations, and seizures; the contraindicated in patients with existing renal
risk of seizures may be increased with insufficiency.
concurrent use of imipenem. Direct intravitreal administration of cidofovir
Foscarnet caused chromosomal damage in is not recommended because of ocular
preclinical studies. toxicity.
The primary adverse effect of intravenous
cidofovir is a dose-dependent proximal
CIDOFOVIR tubular nephrotoxicity, which may be reduced
Cidofovir (eFigure 49–2.1) is a cytosine with prehydration using normal saline.
nucleotide analog with in vitro activity Proteinuria, azotemia, metabolic acidosis, and
against CMV, HSV-1, HSV-2, VZV, EBV, Fanconi’s syndrome may occur.
HHV-6, HHV-8, adenovirus, poxviruses, Concurrent administration of other potentially
polyomaviruses, and human papillomavirus. nephrotoxic agents (eg, amphotericin B,
In contrast to ganciclovir, phosphorylation of aminoglycosides, nonsteroidal anti-
cidofovir to the active diphosphate is inflammatory drugs, pentamidine, foscarnet)
independent of viral enzymes (Figure 49–2); should be avoided.
thus activity is maintained against thymidine Prior administration of foscarnet may increase
kinase-deficient or -altered strains of CMV or the risk of nephrotoxicity.
HSV. Other potential adverse effects include uveitis,
Cidofovir diphosphate acts both as a potent ocular hypotony, and neutropenia (15– 24%).
inhibitor of and as an alternative substrate for Concurrent probenecid use may result in other
viral DNA polymerase, competitively toxicities or drug-drug interactions (see
inhibiting DNA synthesis and becoming Chapter 36).
incorporated into the viral DNA chain. Cidofovir is mutagenic, gonadotoxic, and
Cidofovir-resistant isolates tend to be cross- embryotoxic, and causes hypospermia and
resistant with ganciclovir but retain mammary adenocarcinomas in animals.
susceptibility to foscarnet.
Although the terminal half-life of cidofovir is
approximately 2.6 hours, the active metabolite ANTIRETROVIRAL AGENTS
cidofovir diphosphate has a prolonged
intracellular half-life of 17–65 hours, thus
Substantial advances have been made in Discussion of antiretroviral agents in this
antiretroviral therapy since the introduction of chapter is specific to HIV-1.
the first agent, zidovudine, in 1987. Patterns of susceptibility of HIV-2 to these
Six classes of antiretroviral agents are agents may vary; however, there is generally
currently available for use: innate resistance to the NNRTIs and lower
nucleoside/nucleotide reverse transcriptase barriers of resistance to NRTIs and PIs; data
inhibitors (NRTIs), non-nucleoside reverse regarding maraviroc are inconclusive.
transcriptase inhibitors (NNRTIs), protease
inhibitors (PIs), fusion inhibitors, CCR5 co-
receptor antagonists (also called entry
inhibitors), and HIV integrase strand transfer
inhibitors (INSTIs) (Table 49–3).
These agents inhibit HIV replication at different
parts of the cycle (Figure 49–3). Structures of
some of these drugs are shown in eFigure 49–
3.1.
Greater knowledge of viral dynamics through
the use of viral load and resistance testing has
made it clear that combination therapy with
maximally potent agents will reduce viral
replication to the lowest possible level, thereby
reducing the number of cumulative mutations
and decreasing the likelihood of emergence of
resistance.
Thus, administration of combination
antiretroviral therapy, typically including at least
three antiretroviral agents with differing
susceptibility patterns, has become the standard
of care.
Viral susceptibility to specific agents varies
among patients and may change with time.
Therefore, such combinations must be chosen
with care and tailored to the individual, as must
changes to a given regimen.
In addition to potency and susceptibility,
important factors in the selection of agents for
any given patient are tolerability, convenience,
and optimization of adherence.
As new agents have become available, several
older ones have had diminished usage, because
of either suboptimal safety or inferior antiviral
efficacy.
Zalcitabine (ddC; dideoxycytidine), for example,
is no longer marketed.
Decrease of the circulating viral load by
antiretroviral therapy is correlated with
enhanced survival as well as decreased
morbidity.
Also, recent evidence suggests that in addition to
providing clinical benefits for the patient, the
use of antiretroviral therapy strongly reduces the
risk for heterosexual HIV transmission.
The K65R/N mutation is associated with
reduced susceptibility to tenofovir, abacavir,
lamivudine, and emtricitabine.
All NRTIs may be associated with
mitochondrial toxicity, probably owing to
inhibition of mitochondrial DNA polymerase
gamma.
Less commonly, lactic acidosis with hepatic
steatosis may occur, which can be fatal.
NRTI treatment should be suspended in the
setting of rapidly rising aminotransferase levels,
progressive hepatomegaly, or metabolic acidosis
of unknown cause.
The thymidine analogs zidovudine and
stavudine may be particularly associated with
dyslipidemia and insulin resistance.
Also, some evidence suggests an increased risk
of myocardial infarction in patients receiving
abacavir; this remains unproven.
NUCLEOSIDE & NUCLEOTIDE REVERSE
TRANSCRIPTASE INHIBITORS (NRTIs)

NRTIs are considered the “backbone” of


antiretroviral therapy and are generally used in
combination with other classes of agents, such
as an NNRTI, PI, or integrase inhibitor.
NRTIs are usually given in pairs, and many are
available as coformulations in order to decrease
pill burden and improve adherence.
However, certain NRTI combinations should be
avoided, due to either drug-drug interactions (eg,
didanosine plus tenofovir; see Table 49–4),
similar resistance patterns (eg, lamivudine plus
emtricitabine) or overlapping toxicities (eg, ABACAVIR
stavudine plus didanosine). Abacavir is a guanosine analog that is well
The NRTIs act by competitive inhibition of absorbed following oral administration (83%)
HIV-1 reverse transcriptase; incorporation into and is unaffected by food.
the growing viral DNA chain causes premature The serum half-life is 1.5 hours. The drug
undergoes hepatic glucuronidation and
chain termination due to inhibition of binding
carboxylation.
with the incoming nucleotide (Figure 49–3).
Since the drug is metabolized by alcohol
Each agent requires intracytoplasmic activation dehydrogenase, serum levels of abacavir may
via phosphorylation by cellular enzymes to the be increased with concurrent alcohol (ie,
triphosphate form. ethanol) ingestion.
Typical resistance mutations include M184V, Cerebrospinal fluid levels are approximately
L74V, D67N, and M41L. Lamivudine or one-third those of plasma.
emtricitabine therapy tends to select rapidly for Abacavir is available in a fixed dose
the M184V mutation in regimens that are not formulation with lamivudine and also with
fully suppressive. zidovudine plus lamivudine.
While the M184V mutation confers reduced High-level resistance to abacavir appears to
susceptibility to abacavir, didanosine, and require at least two or three concomitant
zalcitabine, its presence may restore phenotypic mutations and thus tends to develop slowly.
Hypersensitivity reactions, occasionally fatal,
susceptibility to zidovudine.
have been reported in up to 8% of patients pancreatitis.
receiving abacavir and may be more severe in Other risk factors for pancreatitis (eg, alcohol
association with once-daily dosing. abuse, hypertriglyceridemia) are relative
Symptoms, which generally occur within the contraindications, and concurrent use of drugs
first 6 weeks of therapy, include fever, with the potential to cause pancreatitis,
fatigue, nausea, vomiting, diarrhea, and including zalcitabine, stavudine, ribavirin, and
abdominal pain. hydroxyurea, should be avoided (Table 49–
Respiratory symptoms such as dyspnea, 3).
pharyngitis, and cough may also be present, The risk of peripheral distal sensory
and skin rash occurs in about 50% of patients. neuropathy, another potential toxicity, may be
The laboratory abnormalities of a mildly increased with concurrent use of stavudine,
elevated serum aminotransferase or creatine isoniazid, vincristine, or ribavirin.
kinase level may be present but are Other reported adverse effects include
nonspecific. diarrhea (particularly with the buffered
Although the syndrome tends to resolve formulation), hepatitis, esophageal ulceration,
quickly with discontinuation of medication, cardiomyopathy, central nervous system
rechallenge with abacavir results in return of toxicity (headache, irritability, insomnia), and
symptoms within hours and may be fatal. hypertriglyceridemia.
Screening for HLA-B*5701 before initiation Due to an increased risk of lactic acidosis and
of abacavir therapy is recommended to hepatic steatosis when combined with
identify patients with a markedly increased stavudine, this combination should be
risk for abacavir-associated hypersensitivity avoided, especially during pregnancy.
reaction. Previously asymptomatic hyperuricemia may
Although the positive predictive value of this precipitate attacks of gout in susceptible
test is only about 50%, it has a negative individuals; concurrent use of allopurinol may
predictive value approaching 100%. increase levels of didanosine.
Other potential adverse events are rash, fever, Reports of retinal changes and optic neuritis
nausea, vomiting, diarrhea, headache, in patients receiving didanosine, particularly
dyspnea, fatigue, and pancreatitis (rare). in adults receiving high doses and in children,
In some studies but not in others, abacavir has mandate periodic retinal examinations.
been associated with a higher risk of Lipoatrophy appears to be more common in
myocardial infarction. patients receiving didanosine or other
Since abacavir may lower methadone levels, thymidine analogs.
patients receiving these two agents The buffer in didanosine tablets interferes
concurrently should be monitored for signs of with absorption of indinavir, delavirdine,
opioid withdrawal and may require an atazanavir, dapsone, itraconazole, and
increased dose of methadone. fluoroquinolone agents; therefore,
administration should be separated in time.
Serum levels of didanosine are increased
DIDANOSINE when co-administered with tenofovir or
Didanosine (ddI) is a synthetic analog of ganciclovir, and are decreased by atazanavir,
deoxyadenosine. delavirdine, ritonavir, tipranavir, and
Oral bioavailability is approximately 40%. methadone (Table 49–4).
Dosing on an empty stomach is optimal, but Didanosine should not be used in combination
buffered formulations are necessary to with ribavirin.
prevent inactivation by gastric acid (Table
49–3).
Cerebrospinal fluid concentrations of the drug EMTRICITABINE
are approximately 20% of serum Emtricitabine (FTC) is a fluorinated analog of
concentrations. lamivudine with a long intracellular half-life
Serum half-life is 1.5 hours, but the (> 24 hours), allowing for once-daily dosing.
intracellular half-life of the activated Oral bioavailability of the capsules is 93%
compound is as long as 20–24 hours. and is unaffected by food, but penetration into
The drug is eliminated by both cellular the cerebrospinal fluid is low.
metabolism and renal excretion. Elimination is by both glomerular filtration
The major clinical toxicity associated with and active tubular secretion.
didanosine therapy is dose-dependent The serum half-life is about 10 hours.
The oral solution, which contains propylene was 0.2.
glycol, is contraindicated in young children, Serum half-life is 2.5 hours, whereas the
pregnant women, patients with renal or intracellular half-life of the triphosphorylated
hepatic failure, and those using metronidazole compound is 11–14 hours.
or disulfiram. Most of the drug is eliminated unchanged in
Also, because of its activity against HBV, the urine.
patients co-infected with HIV and HBV Lamivudine remains one of the recommended
should be closely monitored if treatment with antiretroviral agents in pregnant women
emtricitabine is interrupted or discontinued, (Table 49–5).
owing to the likelihood of hepatitis flare. Lamivudine is available in a fixed-dose
Emtricitabine is available in a fixed-dose formulation with zidovudine and also with
formulation with tenofovir, either alone or in abacavir.
combination with efavirenz, rilpivirine, or Potential adverse effects are headache,
elvitegravir plus cobicistat (a boosting agent). dizziness, insomnia, fatigue, dry mouth, and
Based on results of clinical trials, the gastrointestinal discomfort, although these are
combination of tenofovir and emtricitabine is typically mild and infrequent.
now recommended as pre-exposure Lamivudine’s bioavailability increases when
prophylaxis to reduce HIV acquisition in men it is co-administered with trimethoprim-
who have sex with men, in heterosexually sulfamethoxazole.
active men and women, and in injection drug Lamivudine and zalcitabine may inhibit the
users. intracellular phosphorylation of one another;
Like lamivudine, the M184V/I mutation is therefore, their concurrent use should be
most frequently associated with emtricitabine avoided if possible.
use and may emerge rapidly in patients
receiving regimens that are not fully
suppressive.
Because of their similar mechanisms of action
and resistance profiles, the combination of
lamivudine and emtricitabine is not
recommended.
The most common adverse effects observed in
patients receiving emtricitabine are headache,
insomnia, nausea, and rash.
In addition, hyperpigmentation of the palms
or soles may be observed (~ 3%), particularly
in African-Americans (up to 13%).

LAMIVUDINE
Lamivudine (3TC) is a cytosine analog STAVUDINE
(eFigure 49–3.1) with in vitro activity against The thymidine analog stavudine (d4T) has
HIV-1 that is synergistic with a variety of high oral bioavailability (86%) that is not
antiretroviral nucleoside analogs—including food-dependent.
zidovudine and stavudine—against both The serum half-life is 1.1 hours, the
zidovudine-sensitive and zidovudine-resistant intracellular half-life is 3.0–3.5 hours, and
HIV-1 strains. mean cerebrospinal fluid concentrations are
As with emtricitabine, lamivudine has activity 55% of those of plasma.
against HBV; therefore, discontinuation in Excretion is by active tubular secretion and
patients that are co-infected with HIV and glomerular filtration.
HBV may be associated with a flareof The major toxicity is a dose-related peripheral
hepatitis. sensory neuropathy.
Lamivudine therapy rapidly selects for the The incidence of neuropathy may be
M184V mutation in regimens that are not increased when stavudine is administered
fully suppressive. with other potentially neurotoxic drugs such
Oral bioavailability exceeds 80% and is not as didanosine, vincristine, isoniazid, or
food-dependent. In children, the average ribavirin, or in patients with advanced
cerebrospinal fluid:plasma ratio of lamivudine
immunosuppression. prophylaxis to reduce HIV acquisition in men
Symptoms typically resolve upon who have sex with men, in heterosexually
discontinuation of stavudine; in such cases, a active men and women, and in injection drug
reduced dosage may be cautiously restarted. users.
Other potential adverse effects are The primary mutations associated with
pancreatitis, arthralgias, and elevation in resistance to tenofovir are K65R/N and K70E.
serum aminotransferases. Gastrointestinal complaints (eg, nausea,
Lactic acidosis with hepatic steatosis, as well diarrhea, vomiting, flatulence) are the most
as lipodystrophy, appear to occur more common adverse effects but rarely require
frequently in patients receiving stavudine than discontinuation of therapy.
in those receiving other NRTI agents. Since tenofovir is formulated with lactose,
Moreover, because the co-administration of these may occur more frequently in patients
stavudine and didanosine may increase the with lactose intolerance.
incidence of lactic acidosis and pancreatitis, Other potential adverse effects include
concurrent use should be avoided. headache, rash, dizziness, and asthenia.
This combination has been implicated in Cumulative loss of renal function has been
several deaths in HIV-infected pregnant observed, possibly increased with concurrent
women. use of boosted PI regimens.
A rare adverse effect is a rapidly progressive Acute renal failure and Fanconi’s syndrome
ascending neuromuscular weakness. have also been reported.
Since zidovudine may reduce the For this reason, tenofovir should be used with
phosphorylation of stavudine, these two drugs caution in patients at risk for renal
should not be used together. dysfunction.
Serum creatinine levels should be monitored
during therapy and tenofovir discontinued for
TENOFOVIR new proteinuria, glycosuria, or calculated
Tenofovir is an acyclic nucleoside glomerular filtration rate < 30 mL/min.
phosphonate (ie, nucleotide) analog of Tenofovir-associated proximal renal
adenosine. tubulopathy causes excessive renal phosphate
Like the nucleoside analogs, tenofovir and calcium losses and 1-hydroxylation
competitively inhibits HIV reverse defects of vitamin D.
transcriptase and causes chain termination Osteomalacia has been demonstrated in
after incorporation into DNA. several animal species, and tenofovir use has
However, only two rather than three been an independent risk factor for bone
intracellular phosphorylations are required for fracture in some studies.
active inhibition of DNA synthesis. Therefore, monitoring of bone mineral
Tenofovir is also approved for the treatment density should be considered with long-term
of patients with HBV infection. use in those with risk factors for (or known)
Tenofovir disoproxil fumarate is a water- osteoporosis, as well as in children;
soluble prodrug of active tenofovir. additionally, alternative agents could be
The oral bioavailability in fasted patients is considered in post-menopausal women.
approximately 25% and increases to 39% Tenofovir may compete with other drugs that
after a high-fat meal. are actively secreted by the kidneys, such as
The prolonged serum (12–17 hours) and cidofovir, acyclovir, and ganciclovir.
intracellular half-lives allow once-daily Concurrent use of atazanavir or
dosing. lopinavir/ritonavir may increase serum levels
Elimination occurs by both glomerular of tenofovir (Table 49–4).
filtration and active tubular secretion, and
dosage adjustment in patients with renal
insufficiency is recommended. ZIDOVUDINE
Tenofovir is available in several fixed-dose Zidovudine (azidothymidine; AZT) is a
formulations with emtricitabine, either alone deoxythymidine analog that is well absorbed
or in combination with efavirenz, rilpivirine, (63%) and distributed to most body tissues
and elvitegravir plus cobicistat. and fluids, including the cerebrospinal fluid,
Based on results of several clinical trials, the where drug levels are 60–65% of those in
combination of tenofovir and emtricitabine is serum.
now recommended as pre-exposure Although the serum half-life averages 1 hour,
the intracellular half-life of the lamivudine, either through inhibition of first-
phosphorylated compound is 3–4 hours, pass metabolism or through decreased
allowing twice-daily dosing. clearance.
Zidovudine is eliminated primarily by renal Zidovudine may decrease phenytoin levels.
excretion following glucuronidation in the Hematologic toxicity may be increased during
liver. co-administration of other myelosuppressive
Zidovudine is available in a fixed-dose drugs such as ganciclovir, ribavirin, and
formulation with lamivudine, either alone or cytotoxic agents.
in combination with abacavir. Combination regimens containing zidovudine
Zidovudine was the first antiretroviral agent and stavudine should be avoided due to in
to be approved and has been well studied. vitro antagonism.
The drug has been shown to decrease the rate
of clinical disease progression and prolong
survival in HIV-infected individuals. NONNUCLEOSIDE REVERSE TRANSCRIPTASE
Efficacy has also been demonstrated in the INHIBITORS (NNRTIs)
treatment of HIV-associated dementia and
thrombocytopenia. The NNRTIs bind directly to HIV-1 reverse
Studies evaluating the use of zidovudine transcriptase (Figure 49–3), resulting in
during pregnancy, labor, and postpartum allosteric inhibition of RNA- and DNA-
showed significant reductions in the rate of dependent DNA polymerase activity.
vertical transmission, and zidovudine remains The binding site of NNRTIs is near to but
one of the first-line agents for use in pregnant
distinct from that of NRTIs.
women (Table 49–5).
High-level zidovudine resistance is generally Unlike the NRTI agents, NNRTIs neither
seen in strains with three or more of the five compete with nucleoside triphosphates nor
most common mutations: M41L, D67N, require phosphorylation to be active.
K70R, T215F, and K219Q. Baseline genotypic testing is recommended prior
However, the emergence of certain mutations to initiating NNRTI treatment because primary
that confer decreased susceptibility to one resistance rates range from approximately 2% to
drug (eg, L74V for didanosine and M184V 8%.
for lamivudine) may enhance zidovudine NNRTI resistance occurs rapidly with
susceptibility in previously zidovudine- monotherapy and can result from a single
resistant strains. mutation.
Withdrawal of zidovudine may permit the The K103N and Y181C mutations confer
reversion of zidovudine-resistant HIV-1 resistance to the first-generation NNRTIs, but
isolates to the susceptible wild-type
not to the newer agents (ie, etravirine,
phenotype.
The most common adverse effect of rilpivirine).
zidovudine is myelosuppression, resulting in Other mutations (eg, L100I, Y188C, G190A)
macrocytic anemia (1–4%) or neutropenia (2– may also confer cross-resistance among the
8%). NNRTI class.
Gastrointestinal intolerance, headaches, and However, there is no cross-resistance between
insomnia may occur but tend to resolve the NNRTIs and the NRTIs; in fact, some
during therapy. nucleoside-resistant viruses display
Lipoatrophy appears to be more common in hypersusceptibility to NNRTIs.
patients receiving zidovudine or other As a class, NNRTI agents tend to be associated
thymidine analogs. with varying levels of gastrointestinal
Less common toxicities include intolerance and skin rash, the latter of which
thrombocytopenia, hyperpigmentation of the
may infrequently be serious (eg, Stevens-
nails, and myopathy.
Johnson syndrome).
High doses can cause anxiety, confusion, and
tremulousness. A further limitation to use of NNRTI agents as a
Increased serum levels of zidovudine may component of antiretroviral therapy is their
occur with concomitant administration of metabolism by the CYP450 system, leading to
probenecid, phenytoin, methadone, innumerable potential drug-drug interactions
fluconazole, atovaquone, valproic acid, and (Tables 49–3 and 49–4).
All NNRTI agents are substrates for CYP3A4 Efavirenz can be given once daily because of
and can act as inducers (nevirapine), inhibitors its long half-life (40–55 hours).
(delavirdine), or mixed inducers and inhibitors It is moderately well absorbed following oral
(efavirenz, etravirine). administration (45%).
Given the large number of non-HIV medications Since toxicity may increase owing to
that are also metabolized by this pathway (see increased bioavailability after a high-fat meal,
efavirenz should be taken on an empty
Chapter 4), drug-drug interactions must be
stomach.
expected and looked for; dosage adjustments are
Efavirenz is principally metabolized by
frequently required and some combinations are CYP3A4 and CYP2B6 to inactive
contraindicated. hydroxylated metabolites; the remainder is
DELAVIRDINE eliminated in the feces as unchanged drug.
Delavirdine has an oral bioavailability of It is highly bound to albumin (~ 99%), and
about 85%, but this is reduced by antacids or cerebrospinal fluid levels range from 0.3% to
H2 -blockers. 1.2% of plasma levels.
It is extensively bound (~ 98%) to plasma The principal adverse effects of efavirenz
proteins and has correspondingly low involve the central nervous system.
cerebrospinal fluid levels. Dizziness, drowsiness, insomnia, nightmares,
Serum half-life is approximately 6 hours. and headache tend to diminish with continued
Skin rash occurs in up to 38% of patients therapy; dosing at bedtime may also be
receiving delavirdine; it typically occurs helpful.
during the first 1–3 weeks of therapy and does Psychiatric symptoms such as depression,
not preclude rechallenge. mania, and psychosis have been observed and
However, severe rash such as erythema may necessitate discontinuation.
multiforme and Stevens-Johnson syndrome Skin rash has also been reported early in
have rarely been reported. therapy in up to 28% of patients; the rash is
Other possible adverse effects are headache, usually mild to moderate in severity and
fatigue, nausea, diarrhea, and increased serum typically resolves despite continuation.
aminotransferase levels. Rarely, rash has been severe or life-
Delavirdine has been shown to be teratogenic threatening.
in rats, causing ventricular septal defects and Other potential adverse reactions are nausea,
other malformations at dosages not unlike vomiting, diarrhea, crystalluria, elevated liver
those achieved in humans. enzymes, and an increase in total serum
Thus, pregnancy should be avoided when cholesterol by 10–20%.
taking delavirdine. High rates of fetal abnormalities, such as
Delavirdine is extensively metabolized by the neural tube defects, occurred in pregnant
CYP3A and CYP2D6 enzymes and also monkeys exposed to efavirenz in doses
inhibits CYP3A4 and 2C9. roughly equivalent to the human dosage;
Therefore, there are numerous potential drug- several cases of congenital anomalies have
drug interactions to consider (Tables 49–3 been reported in humans.
and 49–4). Therefore, efavirenz should be avoided in
The concurrent use of delavirdine with pregnant women, particularly in the first
fosamprenavir and rifabutin is not trimester.
recommended because of decreased As both an inducer and an inhibitor of
delavirdine levels. CYP3A4, efavirenz induces its own
Other medications likely to alter delavirdine metabolism and interacts with the metabolism
levels include didanosine, lopinavir, of many other drugs (Tables 49–3 and 49–4).
nelfinavir, and ritonavir. Since efavirenz may lower methadone levels,
Co-administration of delavirdine with patients receiving these two agents
indinavir or saquinavir prolongs the concurrently should be monitored for signs of
elimination half-life of these protease opioid withdrawal and may require an
inhibitors, thus allowing them to be dosed increased dose of methadone.
twice rather than thrice daily.
ETRAVIRINE
EFAVIRENZ Etravirine was designed to be effective
against strains of HIV that had developed
resistance to first-generation NNRTIs, due to and followed by a 2 mg/kg oral dose to the
mutations such as K103N and Y181C, and is neonate within 3 days after delivery, and
recommended for treatment-experienced nevirapine remains one of the recommended
patients that have resistance to other NNRTIs. agents in pregnant women (Table 49–5).
Although etravirine has a higher genetic There is no evidence of human teratogenicity.
barrier to resistance than the other NNRTIs, However, resistance has been documented
mutations selected by etravirine usually are after this single dose.
associated with resistance to efavirenz, Rash, usually a maculopapular eruption that
nevirapine, and delavirdine. spares the palms and soles, occurs in up to
Etravirine should be taken with a meal to 20% of patients, usually in the first 4–6 weeks
increase systemic exposure. of therapy.
It is highly protein-bound and is primarily Although typically mild and self-limited, rash
metabolized by the liver. is dose-limiting in about 7% of patients.
Mean terminal half-life is approximately 41 Women appear to have an increased incidence
hours. of rash.
The most common adverse effects of When initiating therapy, gradual dose
etravirine are rash, nausea, and diarrhea. escalation over 14 days is recommended to
The rash is typically mild and usually resolves decrease the incidence of rash.
after 1–2 weeks without discontinuation of Severe and life-threatening skin rashes have
therapy. been rarely reported, including Stevens-
Rarely, rash has been severe or life- Johnson syndrome and toxic epidermal
threatening. necrolysis.
Laboratory abnormalities include elevations Nevirapine therapy should be immediately
in serum cholesterol, triglyceride, glucose, discontinued in patients with severe rash and
and hepatic transaminase levels. in those with accompanying constitutional
Transaminase elevations are more common in symptoms; since rash may accompany
patients with HBV or HCV co-infection. hepatotoxicity, liver tests should be assessed.
Etravirine is a substrate as well as an inducer Symptomatic liver toxicity may occur in up to
of CYP3A4 and an inhibitor of CYP2C9 and 4% of patients, may be severe, and is more
CYP2C19; it has many therapeutically frequent in those with higher pretherapy CD4
significant drug-drug interactions (Tables 49– cell counts (ie, > 250 cells/mm3 in women
3 and 49–4). and > 400 cells/mm3 in men), in women, and
Some of the interactions are difficult to in those with HBV or HCV co-infection.
predict. Fulminant, life-threatening hepatitis has been
For example, etravirine may decrease reported, typically within the first 18 weeks of
itraconazole and ketoconazole concentrations therapy.
but increase voriconazole concentrations. Other adverse effects include fever, nausea,
Etravirine should not be given with other headache, and somnolence.
NNRTIs, unboosted protease inhibitors, Nevirapine is a moderate inducer of CYP3A
atazanavir/ritonavir, fosamprenavir/ritonavir, metabolism, resulting in decreased levels of
or tipranavir/ritonavir. amprenavir, indinavir, lopinavir, saquinavir,
efavirenz, and methadone.
Drugs that induce the CYP3A system, such as
NEVIRAPINE rifampin, rifabutin, and St. John’s wort, can
The oral bioavailability of nevirapine is decrease levels of nevirapine, whereas those
excellent (> 90%) and is not food-dependent. that inhibit CYP3A activity, such as
The drug is highly lipophilic and achieves fluconazole, ketoconazole, and
cerebrospinal fluid levels that are 45% of clarithromycin, can increase nevirapine levels.
those in plasma. Since nevirapine may lower methadone
Serum half-life is 25–30 hours. levels, patients receiving these two agents
It is extensively metabolized by the CYP3A concurrently should be monitored for signs of
isoform to hydroxylated metabolites and then opioid withdrawal and may require an
excreted, primarily in the urine. increased dose of methadone.
A single dose of nevirapine (200 mg) is
effective in the prevention of transmission of
HIV from mother to newborn when RILPIVIRINE
administered to women at the onset of labor Rilpivirine is recommended only in treatment-
naive patients with HIV-1 RNA ≤100,000 By preventing post-translational cleavage of the
copies/mL, and only in combination with at Gag-Pol polyprotein, protease inhibitors (PIs)
least 2 other antiretroviral agents. prevent the processing of viral proteins into
It is available in a fixed dose formulation with functional conformations, resulting in the
emtricitabine and tenofovir. production of immature, noninfectious viral
Rilpivirine must be administered with a meal particles (Figure 49–3).
(preferably high fat or > 400 kcal). Unlike the NRTIs, PIs do not need intracellular
Its oral bioavailability can be significantly
activation.
reduced in the presence of acid lowering
agents. Specific genotypic alterations that confer
It should be used with caution with antacids phenotypic resistance are fairly common with
and H2 -receptor antagonists. these agents, thus contraindicating monotherapy.
Rilpivirine use with proton-pump inhibitors Some of the most common mutations conferring
(PPIs) is contraindicated. broad resistance to PIs are substitutions at the
The drug is highly protein bound and the 10, 46, 54, 82, 84, and 90 codons; the number of
terminal elimination half-life is 50 hours. mutations may predict the level of phenotypic
The E138K substitution emerged most resistance. The I50L substitution emerging
frequently during rilpivirine treatment, during atazanavir therapy has been associated
commonly in combination with the M184I with increased susceptibility to other PIs.
substitution. There is cross-resistance with Darunavir and tipranavir appear to have
other NNRTIs, and the combination of
improved virologic activity in patients harboring
rilpivirine with other NNRTIs is not
HIV-1 resistant to other PIs.
recommended.
Rilpivirine is primarily metabolized by As a class, PIs are associated with mild-to-
CYP3A4, and drugs that induce or inhibit moderate nausea, diarrhea, and dyslipidemia.
CYP3A4 may thus affect the clearance of A syndrome of redistribution and accumulation
rilpivirine. of body fat that results in central obesity,
However, clinically significant drug-drug dorsocervical fat enlargement (buffalo hump),
interactions with other antiretroviral agents peripheral and facial wasting, breast
have not been identified to date. enlargement, and a cushingoid appearance has
Concurrent use of carbamazepine, been observed, perhaps less commonly with
dexamethasone, phenobarbital, phenytoin, atazanavir (see below).
proton pump inhibitors, rifabutin, rifampin, Concurrent increases in triglyceride and low-
rifapentine, and St John’s wort is density lipoprotein levels, along with
contraindicated.
hyperglycemia and insulin resistance, have also
Methadone withdrawal may be precipitated
been noted.
with concurrent usage.
The most common adverse effects associated Abacavir, lopinavir/ritonavir, and
with rilpivirine therapy are rash, depression, fosamprenavir/ritonavir have been associated
headache, insomnia, and increased serum with an increased risk of cardiovascular disease
aminotransferases. Increased serum in some, but not all, studies.
cholesterol, and fat redistribution syndrome All PIs may be associated with cardiac
have been reported. Higher doses have been conduction abnormalities, including PR or QT
associated with QTc prolongation. interval prolongation or both.
A baseline electrocardiogram and avoidance of
other agents causing prolonged PR or QT
PROTEASE INHIBITORS (PIs)
intervals should be considered.
Drug-induced hepatitis and rare severe
During the later stages of the HIV growth cycle,
hepatotoxicity have been reported to varying
thegag and gag-pol gene products are translated
degrees with all PIs; the frequency of hepatic
into polyproteins, and these become immature
events is higher with tipranavir/ritonavir than
budding particles.
with other PIs.
The HIV protease is responsible for cleaving
Whether PI agents are associated with bone loss
these precursor molecules to produce the final
and osteoporosis after long-term use is under
structural proteins of the mature virion core.
investigation.
PIs have been associated with increased with various known PI mutations as well as
spontaneous bleeding in patients with with the novel I50L substitution.
hemophilia A or B; an increased risk of Whereas some atazanavir resistance mutations
intracranial hemorrhage has been reported in have been associated in vitro with decreased
patients receiving tipranavir with ritonavir. susceptibility to other PIs, the I50L mutation
All of the antiretroviral PIs are extensively has been associated with increased
susceptibility to other PIs.
metabolized by CYP3A4, with ritonavir having
The most common adverse effects in patients
the most pronounced inhibitory effect and
receiving atazanavir are diarrhea and nausea;
saquinavir the least. vomiting, abdominal pain, headache,
Some PI agents, such as amprenavir and peripheral neuropathy, and skin rash may also
ritonavir, are also inducers of specific CYP occur.
isoforms. As with indinavir, indirect hyperbilirubinemia
As a result, there is enormous potential for drug- with overt jaundice may occur in
drug interactions with other antiretroviral agents approximately 10% of patients, owing to
and other commonly used medications (Tables inhibition of the UGT1A1 glucuronidation
49–3 and 49–4). enzyme.
Expert resources about drug-drug interactions Elevation of hepatic enzymes has also been
should be consulted, as dosage adjustments are observed, usually in patients with underlying
frequently required and some combinations are HBV or HCV co-infection.
Nephrolithiasis has been described in
contraindicated.
association with atazanavir use, and
It is noteworthy that the potent CYP3A4
prolonged use of boosted atazanavir is
inhibitory properties of ritonavir are used to associated with cumulative loss of renal
clinical advantage by having it “boost” the levels function.
of other PI agents when given in combination, In contrast to the other PIs, atazanavir does
thus acting as a pharmacokinetic enhancer rather not appear to be associated with dyslipidemia,
than an antiretroviral agent. fat redistribution, or the metabolic syndrome.
Ritonavir boosting increases drug exposure, As an inhibitor of CYP3A4 and CYP2C9, the
thereby prolonging the drug’s half-life and potential for drug-drug interactions with
allowing reduction in frequency; in addition, the atazanavir is great T( ables 49–3 and 49–4).
genetic barrier to resistance is raised. Atazanavir AUC is reduced by up to 76%
when combined with a proton pump inhibitor;
ATAZANAVIR thus, this combination is to be avoided.
Atazanavir (eFigure 49–3.1) is an azapeptide In addition, co-administration of atazanavir
PI with a pharmacokinetic profile that allows with other drugs that inhibit UGT1A1, such
once-daily dosing. as irinotecan, may increase its levels.
Atazanavir requires an acidic medium for Tenofovir and efavirenz should not be co-
absorption and exhibits pH-dependent administered with atazanavir unless ritonavir
aqueous solubility; therefore, it should be is added to boost levels.
taken with meals and separation of ingestion
from acid-reducing agents by at least 12 hours
is recommended; concurrent proton pump DARUNAVIR
inhibitors are contraindicated. Darunavir is licensed as a PI that must be co-
Atazanavir is able to penetrate both the administered with ritonavir.
cerebrospinal and seminal fluids. Darunavir should be taken with meals to
The plasma half-life is 6–7 hours, which improve bioavailability.
increases to approximately 11 hours when co- It is highly protein-bound and primarily
administered with ritonavir. metabolized by the liver.
The primary route of elimination is biliary; Symptomatic adverse effects of darunavir
atazanavir should not be given to patients include diarrhea, nausea, headache, and rash.
with severe hepatic insufficiency. Laboratory abnormalities include
Atazanavir is one of the recommended dyslipidemia (though possibly less frequent
antiretroviral agents for pregnant women than with other boosted PI regimens) and
(Table 49–5). increases in amylase and hepatic transaminase
Resistance to atazanavir has been associated levels.
Liver toxicity, including severe hepatitis, has
been reported in some patients taking Because the oral solution also contains
darunavir; the risk of hepatotoxicity may be vitamin E at several times the recommended
higher for persons with HBV, HCV, or other daily dosage, supplemental vitamin E should
chronic liver disease. be avoided.
Darunavir contains a sulfonamide moiety and Amprenavir, a sulfonamide, is contraindicated
may cause a hypersensitivity reaction, in patients with a history of sulfa allergy.
particularly in patients with sulfa allergy. Lopinavir/ritonavir should not be co-
Darunavir both inhibits and is metabolized by administered with amprenavir owing to
the CYP3A enzyme system, conferring many decreased amprenavir and altered lopinavir
possible drug-drug interactions (Tables 49–3 exposures.
and 49–4). An increased dosage of amprenavir is
In addition, the co-administered ritonavir is a recommended when co-administered with
potent inhibitor of CYP3A and CYP2D6, and efavirenz (with or without the addition of
an inducer of other hepatic enzyme systems. ritonavir to boost levels).

FOSAMPRENAVIR INDINAVIR
Fosamprenavir is a prodrug of amprenavir Indinavir requires an acidic environment for
that is rapidly hydrolyzed by enzymes in the optimum solubility and therefore must be
intestinal epithelium. consumed on an empty stomach or with a
Because of its significantly lower daily pill small, low-fat, low-protein meal for maximal
burden, fosamprenavir tablets have replaced absorption (60–65%).
amprenavir capsules for adults. The serum half-life is 1.5–2 hours, protein
Fosamprenavir is most often administered in binding is approximately 60%, and the drug
combination with low-dose ritonavir. has a high level of cerebrospinal fluid
After hydrolysis of fosamprenavir, penetration (up to 76% of serum levels).
amprenavir is rapidly absorbed from the Excretion is primarily fecal.
gastrointestinal tract, and its prodrug can be An increase in AUC by 60% and in half-life
taken with or without food. to 2.8 hours in the setting of hepatic
However, high-fat meals decrease absorption insufficiency necessitates dose reduction.
and thus should be avoided. The most common adverse effects of
The plasma half-life is relatively long (7–11 indinavir are indirect hyperbilirubinemia and
hours). nephrolithiasis due to urinary crystallization
Amprenavir is metabolized in the liver and of the drug.
should be used with caution in the setting of Nephrolithiasis can occur within days after
hepatic insufficiency. initiating therapy, with an estimated incidence
The most common adverse effects of of approximately 10%.
fosamprenavir are headache, nausea, diarrhea, Consumption of at least 48 ounces of water
perioral paresthesias, depression. daily is important to maintain adequate
Fosamprenavir contains a sulfa moiety and hydration.
may cause a rash in up to 3% of patients, Thrombocytopenia, elevations of serum
sometimes severe enough to warrant drug aminotransferase levels, nausea, diarrhea,
discontinuation. insomnia, dry throat, dry skin, and indirect
Amprenavir is both an inducer and an hyperbilirubinemia have also been reported.
inhibitor of CYP3A4 and is contraindicated Insulin resistance may be more common with
with numerous drugs (Tables 49–3 and 49–4). indinavir than with the other PIs, occurring in
The oral solution, which contains propylene 3–5% of patients.
glycol, is contraindicated in young children, There have also been rare cases of acute
pregnant women, patients with renal or hemolytic anemia. Since indinavir is an
hepatic failure, and those using metronidazole inhibitor of CYP3A4, numerous and complex
or disulfiram. drug interactions can occur (Tables 49–3 and
Also, the oral solutions of amprenavir and 49–4).
ritonavir should not be co-administered Combination with ritonavir (boosting) allows
because the propylene glycol in one and the for twice-daily rather than thrice-daily dosing
ethanol in the other may compete for the same and eliminates the food restriction associated
metabolic pathway, leading to accumulation with use of indinavir.
of either. However, there is potential for an increase in
nephrolithiasis with this combination The plasma half-life in humans is 3.5–5
compared with indinavir alone; thus, a high hours, and the drug is more than 98% protein-
fluid intake (1.5–2 L/d) is advised. bound.
The most common adverse effects associated
with nelfinavir are diarrhea and flatulence.
LOPINAVIR Diarrhea often responds to antidiarrheal
Lopinavir is currently available only in medications but can be dose-limiting.
combination with ritonavir, which inhibits the Nelfinavir is an inhibitor of the CYP3A
CYP3A-mediated metabolism of lopinavir, system, and multiple drug interactions may
thereby resulting in increased exposure to occur (Tables 49–3 and 49–4).
lopinavir. An increased dosage of nelfinavir is
In addition to improved patient compliance recommended when co-administered with
due to reduced pill burden, lopinavir/ritonavir rifabutin (with a decreased dose of rifabutin),
is generally well tolerated. whereas a decrease in saquinavir dose is
Lopinavir is highly protein bound (98–99%), suggested with concurrent nelfinavir.
and its half-life is 5–6 hours. It is extensively Co-administration with efavirenz should be
metabolized by CYP3A, which is inhibited by avoided due to decreased nelfinavir levels.
ritonavir.
Serum levels of lopinavir may be increased in
patients with hepatic impairment. RITONAVIR
Lopinavir/ritonavir is one of the Ritonavir (eFigure 49–3.1) has a high
recommended antiretroviral agents for use in bioavailability (about 75%) that increases
pregnant women (Table 49–5). with food. It is 98% protein-bound and has a
The most common adverse effects of serum half-life of 3–5 hours.
lopinavir are diarrhea, abdominal pain, Metabolism to an active metabolite occurs via
nausea, vomiting, and asthenia. the CYP3A and CYP2D6 isoforms; excretion
Ritonavir-boosted lopinavir may be more is primarily in the feces.
commonly associated with gastrointestinal Caution is advised when administering the
adverse events than other PIs. drug to persons with impaired hepatic
Elevations in serum cholesterol and function.
triglycerides are common. Ritonavir is one of the recommended
Prolonged use of boosted lopinavir is antiretroviral agents for use in pregnant
associated with cumulative loss of renal women (Table 49–5).
function, and lopinavir use has been an Potential adverse effects of ritonavir,
independent risk factor for bone fracture in particularly when administered at full dosage,
some (but not all) studies. are gastrointestinal disturbances, paresthesias
Potential drug-drug interactions are extensive (circumoral or peripheral), elevated serum
(Tables 49–3 and 49–4). aminotransferase levels, altered taste,
Increased dosage of lopinavir/ritonavir is headache, and elevations in serum creatine
recommended when co-administered with kinase.
efavirenz or nevirapine, which induce Nausea, vomiting, diarrhea, or abdominal pain
lopinavir metabolism. typically occur during the first few weeks of
Concurrent use of fosamprenavir should be therapy but may diminish over time or if the
avoided owing to altered exposure to drug is taken with meals.
lopinavir with decreased levels of amprenavir. Dose escalation over 1–2 weeks is
Also, concomitant use of lopinavir/ritonavir recommended to decrease the dose-limiting
and rifampin is contraindicated due to an side effects.
increased risk for hepatotoxicity. Ritonavir is a potent inhibitor of CYP3A4,
Since the oral solution of lopinavir/ritonavir resulting in many potential drug interactions
contains alcohol, concurrent disulfiram and (Tables 49–3 and 49–4).
metronidazole are contraindicated. However, this characteristic has been used to
great advantage when ritonavir is
administered in low doses (100–200 mg twice
NELFINAVIR daily) in combination with any of the other PI
Nelfinavir has high absorption in the fed state agents, in that increased blood levels of the
(70–80%), undergoes metabolism by CYP3A, latter agents permit lower or less frequent
and is excreted primarily in the feces. dosing (or both) with greater tolerability as
well as the potential for greater efficacy is co-administered with delavirdine or
against resistant virus. rifampin.
Therapeutic levels of digoxin and
theophylline should be monitored when co-
administered with ritonavir owing to a likely TIPRANAVIR
increase in their concentrations. Tipranavir is a newer PI indicated for use in
The concurrent use of saquinavir and ritonavir treatment-experienced patients who harbor
is contraindicated due to an increased risk of strains resistant to other PI agents.
QT prolongation (with torsades de pointes It is used in combination with ritonavir to
arrhythmia) and PR interval prolongation. achieve effective serum levels.
Bioavailability is poor but is increased when
taken with a high-fat meal.
SAQUINAVIR The drug is metabolized by the liver
In its original formulation as a hard gel microsomal system and is contraindicated in
capsule, oral saquinavir was poorly patients with hepatic insufficiency.
bioavailable (only about 4% after food). Tipranavir contains a sulfonamide moiety and
However, reformulation of saquinavir for should not be administered to patients with
once-daily dosing in combination with low- known sulfa allergy.
dose ritonavir has both improved antiviral The most common adverse effects of
efficacy and decreased gastrointestinal tipranavir are diarrhea, nausea, vomiting, and
adverse effects. abdominal pain.
A previous formulation of saquinavir in soft An urticarial or maculopapular rash is more
gel capsules is no longer available. common in women and may be accompanied
Saquinavir should be taken within 2 hours by systemic symptoms or desquamation.
after a fatty meal for enhanced absorption. Liver toxicity, including life-threatening
Saquinavir is 97% protein-bound, and serum hepatic decompensation, has been observed
half-life is approximately 2 hours. and may be more common than with other
Saquinavir has a large volume of distribution, PIs, particularly in patients with chronic HBV
but penetration into the cerebrospinal fluid is or HCV infection.
negligible. Tipranavir should be discontinued in patients
Excretion is primarily in the feces. who have increased serum transaminase
Reported adverse effects include levels that are more than 10 times the upper
gastrointestinal discomfort (nausea, diarrhea, limit of normal or more than 5 times normal
abdominal discomfort, dyspepsia) and in combination with increased serum
rhinitis. bilirubin.
When administered in combination with low- Because of an increased risk for intracranial
dose ritonavir, there appears to be less hemorrhage in patients receiving
dyslipidemia or gastrointestinal toxicity than tipranavir/ritonavir, the drug should be
with some of the other boosted PI regimens. avoided in patients with head trauma or
However, the concurrent use of saquinavir bleeding diathesis.
and ritonavir may confer an increased risk of Other potential adverse effects include
QT prolongation (with torsades de pointes depression, elevation in amylase, and
arrhythmia) and PR interval prolongation. decreased white blood cell count.
Saquinavir is subject to extensive first-pass Tipranavir both inhibits and induces the
metabolism by CYP3A4 and functions as a CYP3A4 system. When used in combination
CYP3A4 inhibitor as well as a substrate; thus, with ritonavir, its net effect is inhibition.
there are many potential drug-drug Tipranavir also induces P-glycoprotein
interactions (Tables 49–3 and 49–4). transporter and thus may alter the disposition
A decreased dose of saquinavir is of many other drugs (Table 49–4). Concurrent
recommended when co-administered with administration of tipranavir with
nelfinavir. Increased saquinavir levels when fosamprenavir or saquinavir should be
co-administered with omeprazole necessitate avoided owing to decreased blood levels of
close monitoring for toxicities. the latter drugs. Tipranavir/ritonavir may also
Digoxin levels may increase if co- decrease serum levels of valproic acid and
administered with saquinavir and should omeprazole. Levels of lovastatin, simvastatin,
therefore be monitored. atorvastatin, and rosuvastatin may be
Liver tests should be monitored if saquinavir increased, increasing the risk for
rhabdomyolysis and myopathy. discontinuation.
Other side effects may include insomnia,
headache, dizziness, and nausea.
ENTRY INHIBITORS Hypersensitivity reactions may rarely occur,
are of varying severity, and may recur on
The process of HIV-1 entry into host cells is rechallenge.
complex; each step presents a potential target for Eosinophilia is the primary laboratory
inhibition. abnormality seen with enfuvirtide
Viral attachment to the host cell entails binding administration.
of the viral envelope glycoprotein complex No drug-drug interactions have been
gp160 (consisting of gp120 and gp41) to its identified that would require the alteration of
the dosage of concomitant antiretroviral or
cellular receptor CD4.
other drugs.
This binding induces conformational changes in
gp120 that enable access to the chemokine
receptors CCR5 or CXCR4. MARAVIROC
Chemokine receptor binding induces further Maraviroc (eFigure 49–3.1) is approved for
conformational changes in gp120, allowing use in combination with other antiretroviral
exposure to gp41 and leading to fusion of the agents in treatment-experienced adult patients
viral envelope with the host cell membrane and infected with only CCR5-tropic HIV-1
subsequent entry of the viral core into the detectable who are resistant to other
cellular cytoplasm. antiretroviral agents.
Maraviroc binds specifically and selectively
to the host protein CCR5, one of two
chemokine receptors necessary for entrance of
ENFUVIRTIDE HIV into CD4+ cells.
Enfuvirtide is a synthetic 36-amino-acid Since maraviroc is active against HIV that
peptide fusion inhibitor that blocks HIV entry uses the CCR5 co-receptor exclusively, and
into the cell (Figure 49–3). not against HIV strains with CXCR4, dual, or
Enfuvirtide binds to the gp41 subunit of the mixed tropism, co-receptor tropism should be
viral envelope glycoprotein, preventing the determined by specific testing before
conformational changes required for the maraviroc is started, using the enhanced
fusion of the viral and cellular membranes. sensitivity tropism assay.
It is administered in combination with other Substantial proportions of patients,
antiretroviral agents in treatment-experienced particularly those with advanced HIV
patients with evidence of viral replication infection, are likely to have virus that is not
despite ongoing antiretroviral therapy. exclusively CCR5-tropic.
Enfuvirtide, which must be administered by The absorption of maraviroc is rapid but
subcutaneous injection, is the only variable, with the time to maximum
parenterally administered antiretroviral agent. absorption generally being 1–4 hours after
Metabolism appears to be by proteolytic ingestion of the drug.
hydrolysis without involvement of the Most of the drug (≥ 75%) is excreted in the
CYP450 system. feces, whereas approximately 20% is excreted
Elimination half-life is 3.8 hours. in urine.
Resistance to enfuvirtide can result from The recommended dose of maraviroc varies
mutations in gp41; the frequency and according to renal function and the
significance of this phenomenon are being concomitant use of CYP3A inducers or
investigated. inhibitors.
However, enfuvirtide lacks cross-resistance Maraviroc is contraindicated in patients with
with the other currently approved severe or end-stage renal impairment who are
antiretroviral drug classes. taking concurrent CYP3A inhibitors or
The most common adverse effects associated inducers, and caution is advised when used in
with enfuvirtide therapy are local injection patients with preexisting hepatic impairment
site reactions, consisting of painful and in those co-infected with HBV or HCV.
erythematous nodules. Maraviroc has excellent penetration into the
Although frequent, these are typically mild- cervicovaginalfluid, with levels almost four
to-moderate and rarely lead to times higher than the corresponding
concentrations in blood plasma. This class of agents binds integrase, a viral
Resistance to maraviroc is associated with enzyme essential to the replication of both HIV-
one or more mutations in the V3 loop of 1 and HIV-2.
gp120. By doing so, it inhibits strand transfer, the third
There appears to be no cross-resistance with and final step of provirus integration, thus
drugs from any other class, including the interfering with the integration of reverse-
fusion inhibitor enfuvirtide. However, transcribed HIV DNA into the chromosomes of
emergence of CXCR4 virus (either previously
host cells (Figure 49–3).
undetected or newly developed) appears to be
a more common cause of virologic failure As a class, these agents tend to be well tolerated,
than the development of resistance mutations. with headache and gastrointestinal effects being
Maraviroc is a substrate for CYP3A4 and the most commonly reported adverse events.
therefore requires adjustment in the presence Other nervous system (including
of drugs that interact with these enzymes neuropsychiatric) effects are often reported but
(Tables 49–3 and 49–4). are milder and less frequent than with efavirenz.
It is also a substrate for P-glycoprotein, which Limited data suggest that effects upon lipid
limits intracellular concentrations of the drug. metabolism are favorable compared with
The dosage of maraviroc must be decreased if efavirenz and PIs, with more variable findings
it is co-administered with strong CYP3A for elvitegravir than raltegravir and dolutegravir
inhibitors (eg, delavirdine, ketoconazole, due to co-administration with the boosting agent
itraconazole, clarithromycin, or any protease
cobicistat.
inhibitor other than tipranavir) and must be
Rare and severe events include systemic
increased if coadministered with CYP3A
inducers (eg, efavirenz, etravirine, rifampin, hypersensitivity reactions and rhabdomyolysis.
carbamazepine, phenytoin, or St. John’s DOLUTEGRAVIR
wort). Dolutegravir may be taken with or without
Potential adverse effects of maraviroc include food.
cough, upper respiratory tract infections, The absolute oral bioavailability has not been
muscle and joint pain, diarrhea, sleep established.
disturbance, and elevations in serum Dolutegravir should be taken 2 hours before
aminotransferases. or 6 hours after taking cation-containing
Hepatotoxicity has been reported, which may antacids or laxatives, sucralfate, oral iron
be preceded by a systemic allergic reaction supplements, oral calcium supplements, or
(ie, pruritic rash, eosinophilia, or elevated buffered medications.
IgE); discontinuation of maraviroc should be The terminal half-life is approximately 14
prompt if this constellation occurs. hours.
Also, caution should be used in patients with Dolutegravir is primarily metabolized via
pre-existing liver dysfunction or who are co- UGT1A1 with some contribution from
infected with HBV or HCV. CYP3A.
Myocardial ischemia and infarction have been Therefore, drug-drug interactions may occur
observed in patients receiving maraviroc; (Table 49–4).
therefore caution is advised in patients at Co-administration with the metabolic
increased cardiovascular risk. inducers phenytoin, phenobarbital,
There has been concern that blockade of the carbamazepine, and St. John’s wort should be
chemokine CCR5 receptor—a human protein avoided.
—may result in decreased immune Dolutegravir inhibits the renal organic cation
surveillance, with a subsequent increased risk transporter OCT2, thereby increasing plasma
of malignancy (eg, lymphoma) or infection. concentrations of drugs eliminated via OCT2
To date, however, there has been no evidence such as dofetilide and metformin.
of an increased risk of either malignancy or For this reason, co-administration with
infection in patients receiving maraviroc. dofetilide is contraindicated and close
monitoring, with potential for dose
INTEGRASE STRAND TRANSFER INHIBITORS adjustment, is recommended for co-
administration with metformin.
(INSTIs)
Current evidence suggests that dolutegravir
retains activity against some viruses resistant
to both raltegravir and elvitegravir. concentrations, the dose of raltegravir should
The most common adverse reactions be increased.
associated with dolutegravir are insomnia and Since polyvalent cations (eg, magnesium,
headache. calcium, and iron) may bind integrase
Hypersensitivity reactions characterized by inhibitors and interfere with their activity,
rash, constitutional findings, and sometimes antacids should be used cautiously and
organ dysfunction, including liver injury, ingestion separated by at least 4 hours from
have been reported and may be life- raltegravir. T
threatening. he chewable tablets may contain
The drug should be discontinued immediately phenylalanine, which can be harmful to
if this occurs and not restarted. patients with phenylketonuria.
Other reported side effects include elevations Although virologic failure has been
in serum aminotransferases and the fat uncommon in clinical trials of raltegravir to
redistribution syndrome. date, in vitro resistance requires only a single
point mutation (eg, at codons 148 or 155).
The low genetic barrier to resistance
ELVITEGRAVIR emphasizes the importance of combination
Elvitegravir requires boosting with an therapies and of adherence.
additional drug, such as cobicistat (a Integrase mutations are not expected to affect
pharmacokinetic enhancer that inhibits sensitivity to other classes of antiretroviral
CYP3A4 as well as certain intestinal transport agents.
proteins) or ritonavir. Potential adverse effects of raltegravir include
Elvitegravir is therefore available only as a insomnia, headache, dizziness, diarrhea,
component of a fixed-dose combination, with nausea, fatigue, and muscle aches. Increases
cobicistat, emtricitabine, and tenofovir. in pancreatic amylase, serum
The combined formulation should be taken aminotransferases, and creatine kinase (with
with food. rhabdomyolysis) may occur.
Cobicistat can inhibit renal tubular secretion Severe, potentially life-threatening and fatal
of creatinine, causing increases in serum skin reactions have been reported, including
creatinine that may not be clinically Stevens-Johnson syndrome, hypersensitivity
significant; in the fixed-dose formulation it reaction, and toxic epidermal necrolysis.
may be difficult to distinguish between
cobicistat effect and tenofovir-induced
nephrotoxicity. ANTIHEPATITIS AGENTS
The recommendation is that the fixed-dose
combination INTERFERON ALFA
elvitegravir/cobicistat/tenofovir/emtricitabine
should not be initiated in patients with Interferons are host cytokines that exert complex
calculated creatinine clearance < 70 mL/min antiviral, immunomodulatory, and
and should be discontinued in those with antiproliferative actions (see Chapter 55) and
creatinine clearance < 50 mL/min; some have proven useful in both HBV and
discontinuation should be considered if the HCV.
serum creatinine increases by 0.4 mg/dL or Interferon alfa appears to function by induction
more. of intracellular signals following binding to
specific cell membrane receptors, resulting in
RALTEGRAVIR inhibition of viral penetration, translation,
Absolute bioavailability of the pyrimidinone transcription, protein processing, maturation,
analog raltegravir has not been established but and release, as well as increased host expression
does not appear to be food-dependent. of major histocompatibility complex antigens,
The drug does not interact with the enhanced phagocytic activity of macrophages,
cytochrome P450 system butis metabolized and augmentation of the proliferation and
by glucuronidation, particularly UGT1A1. survival of cytotoxic T cells.
Inducers or inhibitors of UGT1A1 may affect Injectable preparations of interferon alfa are
serum levels of raltegravir. available for treatment of both HBV and HCV
For example, since concurrent use of rifampin
infections (Table 49–6).
substantially decreases raltegravir
Interferon alfa-2a and interferon alfa-2b may be Co-administration with didanosine is not
administered either subcutaneously or recommended because of a risk of hepatic
intramuscularly; half-life is 2–5 hours, failure, and co-administration with zidovudine
depending on the route of administration. may exacerbate cytopenias.
Alfa interferons are filtered at the glomerulus The polyethylene glycol molecule is a nontoxic
and undergo rapid proteolytic degradation polymer that is readily excreted in the urine.
during tubular reabsorption, such that detection
TREATMENT OF HEPATITIS B VIRUS
in the systemic circulation is negligible.
INFECTION
Liver metabolism and subsequent biliary
excretion are considered minor pathways. The goals of chronic HBV therapy are the
The use of pegylated (polyethylene glycol- suppression of HBV DNA to undetectable
complexed) interferon alfa-2a and pegylated levels, seroconversion of HBeAg (or more
interferon alfa-2b results in slower clearance, rarely, HBsAg) from positive to negative, and
longer terminal half-lives, and steadier reduction in elevated hepatic transaminase
concentrations, thus allowing for less frequent levels.
dosing. These end points are correlated with
Renal elimination accounts for about 30% of improvement in necroinflammatory disease, a
clearance, and clearance is approximately halved decreased risk of hepatocellular carcinoma and
in subjects with impaired renal function; dosage cirrhosis, and a decreased need for liver
must therefore be adjusted. transplantation.
The adverse effects of interferon alfa include a All of the currently licensed therapies achieve
flu-like syndrome (ie, headache, fevers, chills, these goals.
myalgias, and malaise) that typically occurs However, because current therapies suppress
within 6 hours after dosing; this syndrome HBV replication without eradicating the virus,
occurs in more than 30% of patients during the initial responses may not be durable.
first week of therapy and tends to resolve upon The covalently closed circular (ccc) viral DNA
continued administration. exists in stable form indefinitely within the cell,
Transient hepatic enzyme elevations may occur serving as a reservoir for HBV throughout the
in the first 8–12 weeks of therapy and appear to life of the cell and resulting in the capacity to
be more common in responders. reactivate.
Potential adverse effects during chronic therapy Relapse is more common in patients co-infected
include neurotoxicities (mood disorders, with HBV and hepatitis D virus.
depression, somnolence, confusion, seizures), As of 2013 seven drugs were approved for
myelosuppression, profound fatigue, weight treatment of chronic HBV infection in the
loss, rash, cough, myalgia, alopecia, tinnitus, United States: five oral nucleoside/nucleotide
reversible hearing loss, retinopathy, analogs (lamivudine, adefovir dipivoxil,
pneumonitis, and possibly cardiotoxicity. tenofovir, entecavir, telbivudine) and two
Induction of autoantibodies may occur, causing injectable interferon drugs (interferon alfa-
exacerbation or unmasking of autoimmune 2b, pegylated interferon alfa-2a) (Table 49–
disease (particularly thyroiditis). 6).
Contraindications to interferon alfa therapy
include hepatic decompensation, autoimmune
disease, and history of cardiac arrhythmia.
Caution is advised in the setting of psychiatric
disease, epilepsy, thyroid disease, ischemic
cardiac disease, severe renal insufficiency, and
cytopenia.
Alfa interferons are abortifacient in primates and
should not be administered in pregnancy.
Potential drug-drug interactions include
The use of interferon has been supplanted by
increased theophylline and methadone levels.
long-acting pegylated interferon, allowing once-
weekly rather than daily or thrice-weekly Protein binding is low (< 5%).
dosing. The intracellular half-life of the diphosphate is
In general, nucleoside/nucleotide analog prolonged, ranging from 5 to 18 hours in various
therapies have better tolerability and produce a cells; this makes once-daily dosing feasible.
higher response rate than the interferons and are Adefovir is excreted by a combination of
now considered the first line of therapy. glomerular filtration and active tubular secretion
Combination therapies may reduce the and requires dose adjustment for renal
development of resistance. dysfunction; however, it may be administered to
The optimal duration of therapy remains patients with decompensated liver disease.
unknown. Several anti-HBV agents have anti- Of the oral agents, adefovir may be slower to
HIV activity as well, including tenofovir, suppress HBV DNA levels and the least likely to
lamivudine, and adefovir dipivoxil. induce HBeAg seroconversion.
Emtricitabine, an NRTI used in HIV infection, Emergence of resistance is 20% to 30% after 5
has resulted in excellent biochemical, virologic, years of use.
and histologic improvement in patients with Naturally occurring (ie, primary) adefovir-
chronic HBV infection, although it is not resistant rt233 HBV mutants have been
approved for this indication. described.
Agents with dual HBV and HIV activity are There is no cross-resistance between adefovir
particularly useful as part of a first-line regimen and lamivudine or entecavir.
in co-infected patients. Adefovir is well tolerated.
However, it is important to note that acute A dose-dependent nephrotoxicity, manifested by
exacerbation of hepatitis may occur upon increased serum creatinine and decreased serum
discontinuation or interruption of these agents; phosphorous, may occur and is more common
this may be severe or even fatal. with use of higher doses (30-60 mg/d) or pre-
existing azotemia.
Other potential adverse effects are headache,
diarrhea, asthenia, and abdominal pain.
As with other NRTI agents, lactic acidosis and
hepatic steatosis are considered a risk owing to
mitochondrial dysfunction.
Pivalic acid, a by-product of adefovir
metabolism, can esterify free carnitine and result
ADEFOVIR DIPIVOXIL in decreased carnitine levels.
However, it is not necessary to administer
Although initially and abortively developed for carnitine supplementation with the low doses
treatment of HIV infection, adefovir dipivoxil used to treat patients with HBV (10 mg/d).
gained approval, at lower and less toxic doses, Severe acute exacerbations of hepatitis have
for treatment of HBV infection. been reported in up to 25% of patients who
Adefovir dipivoxil is the diester prodrug of discontinued adefovir.
adefovir, an acyclic phosphonated adenine Adefovir is embryotoxic in rats at high doses
nucleotide analog (eFigure 49–4.1). and is genotoxic in preclinical studies.
It is phosphorylated by cellular kinases to the
active diphosphate metabolite and then ENTECAVIR
competitively inhibits HBV DNA polymerase Entecavir is an orally administered guanosine
and causes chain termination after incorporation nucleoside analog. that competitively inhibits
into viral DNA. all three functions of HBV DNA polymerase,
including base priming, reverse transcription
Adefovir is active in vitro against a wide range
of the negative strand, and synthesis of the
of DNA and RNA viruses, including HBV, HIV,
positive strand of HBV DNA.
and herpesviruses. Oral bioavailability approaches 100% but is
Oral bioavailability of adefovir dipivoxil is decreased by food; therefore, entecavir should
about 59% and is unaffected by meals; it is be taken on an empty stomach.
rapidly and completely hydrolyzed to the parent The intracellular half-life of the active
compound by intestinal and blood esterases.
phosphorylated compound is 15 hours and with deoxycytidine triphosphate for
plasma half-life is prolonged at 128-149 incorporation into the viral DNA, resulting in
hours, allowing once-daily dosing. chain termination.
It is excreted by the kidney, undergoing both Although lamivudine initially results in rapid
glomerular filtration and net tubular secretion. and potent virus suppression, chronic therapy
Suppression of HBV DNA levels was greater is limited by the emergence of lamivudine-
with entecavir than with lamivudine or resistant HBV isolates (eg, L180M or
adefovir in comparative trials Entecavir M204I/V), estimated to occur in 15–30% of
appears to have a higher barrier to the patients at 1 year and 70% at 5 years of
emergence of resistance than lamivudine but therapy.
resistance may be more likely in the setting of Resistance has been associated with flares of
lamivudine resistance. hepatitis and progressive liver disease.
Although selection of resistant isolates with Cross-resistance between lamivudine and
the S202G mutation has been documented emtricitabine or entecavir may occur;
during therapy, clinical resistance is rare (< however, adefovir and tenofovir maintain
1% at 4 years). activity against lamivudine-resistant strains of
Entecavir has weak anti-HIV activity and can HBV.
induce development of the M184V variant in In the doses used for HBV infection,
HBV/HIV co-infected patients, resulting in lamivudine has an excellent safety profile.
resistance to emtricitabine and lamivudine. Headache, nausea, diarrhea, dizziness,
Entecavir is well tolerated. myalgia, and malaise are rare.
Potential adverse events are headache, Co-infection with HIV may increase the risk
fatigue, dizziness, nausea, rash, and fever. of pancreatitis.
Lung adenomas and carcinomas in mice,
hepatic adenomas and carcinomas in rats and
mice, vascular tumors in mice, and brain TELBIVUDINE
gliomas and skin fibromas in rats have been Telbivudine is a thymidine nucleoside analog
observed at varying exposures. with activity against HBV DNA polymerase.
Co-administration of entecavir with drugs that It is phosphorylated by cellular kinases to the
reduce renal function or compete for active active triphosphate form, which has an
tubular secretion may increase serum intracellular half-life of 14 hours.
concentrations of either entecavir or the co- The phosphorylated compound competitively
administered drug. inhibits HBV DNA polymerase, resulting in
incorporation into viral DNA and chain
termination.
LAMIVUDINE It is not active in vitro against HIV-1. Oral
The pharmacokinetics of lamivudine are bioavailability is unaffected by food.
described earlier in this chapter (see section, Plasma protein-binding is low (3%) and
Nucleoside and Nucleotide Reverse distribution wide.
Transcriptase Inhibitors). The serum half-life is approximately 15 hours
The more prolonged intracellular half-life in and excretion is renal.
HBV cell lines (17–19 hours) than in HIV- There are no known metabolites and no
infected cell lines (10.5–15.5 hours) allows known interactions with the CYP450 system
for lower doses and less frequent or other drugs.
administration. Telbivudine induced greater rates of virologic
Lamivudine can be safely administered to response than either lamivudine or adefovir in
patients with decompensated liver disease. comparative trials.
Prolonged treatment has been shown to However, emergence of resistance, typically
decrease clinical progression of HBV, as well due to the M204I mutation, may occur in up
as development of hepatocellular cancer by to 22% of patients with durations of therapy
approximately 50%. exceeding 1 year, and may result in virologic
Also, lamivudine has been effective in rebound. Telbivudine is not effective in
preventing vertical transmission of HBV from patients with lamivudine-resistant HBV.
mother to newborn when given in the last 4 Adverse effects are mild; they include fatigue,
weeks of gestation. headache, cough, nausea, diarrhea, rash, and
Lamivudine inhibits HBV DNA polymerase fever.
and HIV reverse transcriptase by competing Both uncomplicated myalgia and myopathy
have been reported, concurrent with increased in a sustained rate of clearance of 95% at 6
creatine kinase levels, as has peripheral months.
neuropathy. Therefore, if HCV RNA testing documents
As with other nucleoside analogs, lactic persistent viremia 12 weeks after initial
acidosis and severe hepatomegaly with seroconversion, antiviral therapy is
steatosis may occur during therapy as well as recommended.
flares of hepatitis after discontinuation. Treatment of patients with chronic HCV
infection is recommended for those with an
TENOFOVIR increased risk for progression to cirrhosis.
Tenofovir, a nucleotide analog of adenosine The parameters for selection are complex. In
in use as an antiretroviral agent, has potent those who are to be treated, the traditional
activity against HBV. standard treatment is once-weekly pegylated
The characteristics of tenofovir are described interferon alfa in combination with daily oral
earlier in this chapter. ribavirin.
Tenofovir maintains activity against Pegylated interferon alfa-2a and -2b have
lamivudine- and entecavir-resistant hepatitis replaced their unmodified interferon alfa
virus isolates but has reduced activity against counterparts because of superior efficacy in
adefovir-resistant strains.
combination with ribavirin, regardless of
Although similar in structure to adefovir
dipivoxil, comparative trials show a higher genotype.
rate of virologic response and histologic It is also clear that combination therapy with
improvement, and a lower rate of emergence oral ribavirin is more effective than
of resistance to tenofovir in patients with monotherapy with either interferon or ribavirin
chronic HBV infection. alone.
The most common adverse effects of Therefore, monotherapy with pegylated
tenofovir in patients with HBV infection are interferon alfa is recommended only in patients
nausea, abdominal pain, diarrhea, dizziness, who cannot tolerate ribavirin.
fatigue, and rash; other potential adverse Interferon plus ribavirin therapy is active against
effects are those listed earlier. all genotypes of HCV infection, with SVR rates
of 70 to 80% among patients with HCV
TREATMENT OF HEPATITIS C INFECTION genotype 2 or 3 infection and rates of 45 to 70%
among patients with any of the other genotypes.
In contrast to the treatment of patients with A genetic variant near the gene encoding
chronic HBV infection, the primary goal of interferon-lambda-3 (IL28B rs12979860) is a
treatment in patients with HCV infection is viral strong predictor of response to peginterferon alfa
eradication. and ribavirin.
In clinical trials, the primary efficacy end point However, the recent advent of NS3/4A protease
is typically achievement of sustained viral inhibitors and the NS5B polymerase inhibitors is
response (SVR), defined as the absence of changing the face of chronic HCV therapy.
detectable viremia 24 weeks after completion of Administration of boceprevir, simeprevir, or
therapy. telaprevir, in combination with peginterferon
SVR is associated with improvement in liver and ribavirin, dramatically increased the rate of
histology, reduction in risk of hepatocellular viral clearance in patients with HCV genotype 1;
carcinoma, and, occasionally, with regression of sofosbuvir is effective against HCV genotypes
cirrhosis as well. 1, 2, 3, and 4.
Late relapse occurs in less than 5% of patients Although all four of these new agents are
who achieve SVR. licensed to be administered in combination with
In acute hepatitis C, the rate of clearance of the peginterferon and ribavirin, recent results of
virus without therapy is estimated at 15–30%. In clinical trials have provided evidence that one or
one (uncontrolled) study, treatment of acute more of them may be effective in interferon- and
infection with interferon alfa-2b, in doses higher ribavirin-free regimens.
than those used for chronic hepatitis C, resulted POLYMERASE INHIBITORS
Sofosbuvir potential decrease in serum levels of the anti-
Sofosbuvir is a nucleotide analog that inhibits HCV agent, and co-administration with statin
the HCV NS5B RNA-dependent RNA agents is contraindicated due to increased serum
polymerase in patients infected with HCV levels of the statin agent.
genotype 1, 2, 3, or 4. The effectiveness of hormonal contraceptives
It is administered once daily, with or without may be reduced by co-administration with
food, in combination with peginterferon alfa boceprevir or telaprevir.
and ribavirin, for a total of 12–24 weeks (the
Since boceprevir, simeprevir, and telaprevir are
longer duration is recommended in patients
infected with HCV genotype 3). always co-administered with ribavirin, their use
Very high cure rates are reported but the drug in pregnant women and in men with pregnant
is extraordinarily expensive. partners is contraindicated.
Sofosbuvir is 61–65% bound to plasma
Boceprevir
proteins and is metabolized in the liver to
Boceprevir therapy is initiated after the
form the active nucleoside analog
administration of peginterferon and ribavirin
triphosphate GS-461203.
therapy for 4 weeks. The duration of therapy
Elimination is by renal clearance, and safety
is dependent on the achievement of
has not been established in patients with
undetectable virus. Boceprevir should be
severe renal insufficiency.
taken with food to maximize absorption. It is
Sofosbuvir is a substrate of drug transporter
~75% protein-bound and has a mean plasma
P-gp; therefore, potent P-gp inducers in the
halflife of approximately 3.4 hours.
intestine should not be co-administered.
Boceprevir is metabolized by the aldo-keto-
Commonly reported adverse effects are
reductase and CYP3A4/5 pathways and is an
fatigue and headache.
inhibitor of CYP3A4/5 and P-glycoprotein
transporter. Co-administration of boceprevir
PROTEASE INHIBITORS with numerous drugs is contraindicated,
including carbamazepine, phenobarbital,
phenytoin, rifampin, ergot derivatives,
Three oral protease NS3/4A inhibitors have
cisapride, lovastatin, simvastatin, St. John’s
recently become available for the treatment of wort, drospirenone, alfuzosin, sildenafil or
HCV genotype 1 infection, in combination with tadalafil when used for pulmonary
peginterferon and ribavirin: boceprevir, hypertension, pimozide, triazolam,
simeprevir, and telaprevir. midazolam, and efavirenz.
These agents inhibit HCV replication directly by The most commonly reported adverse effects
binding to the NS3/4A protease that cleaves associated with boceprevir therapy are
HCV-encoded polyproteins (Figure 49–4). fatigue, anemia, neutropenia, nausea,
Of concern is the enhanced toxicity when used headache, and dysgeusia.
in combination with peginterferon and ribavirin, Rates of anemia are higher in patients taking
the high potential for drug-drug interactions, and boceprevir with peginterferon and ribavirin
the low genetic barrier to resistance, which may than in those taking peginterferon and
ribavirin alone (~ 50% vs 25%, respectively);
develop as early as 4 days after initiation of
rates of neutropenia are also higher.
therapy when administered as monotherapy.
Use of these agents in the treatment of other
HCV genotypes is not recommended. Simeprevir
Cross-resistance is expected among NS3/4A Simeprevir is administered once daily in
protease inhibitors. combination with peginterferon and ribavirin
All three agents are inhibitors and substrates of for a total of 12 weeks in patients with
CYP3A inhibitors. compensated liver disease (including
Drug-drug interactions are to be expected with cirrhosis) that are infected with HCV
many concurrent agents, particularly the genotype 1.
Simeprevir must be taken with food to
NNRTIs and PIs in patients with HIV/HCV co-
maximize absorption.
infection.
It is extensively bound to plasma proteins (>
Co-administration with strong CYP3A4 inducers 99%), metabolized in the liver by CYP3A
(including rifampin) is contraindicated due to pathways, and undergoes biliary excretion.
Its safety in patients with moderate to severe associated with telaprevir therapy are rash
liver insufficiency has not been established. (30–55%), anemia, fatigue, pruritus, nausea,
Mean simeprevir exposures are more than and anorectal discomfort.
threefold higher in Asian patients compared Severe rash or Stevens-Johnson syndrome has
with Caucasians, leading to potentially higher been reported; in these patients, the drug
frequencies of adverse events. should be stopped and not restarted.
Simeprevir is a substrate and mild inhibitor of Rates of anemia are higher in patients taking
CYP3A and a substrate and inhibitor of P-gp telaprevir with peginterferon and ribavirin
and OATP1B1/3. than in those taking peginterferon and
Co-administration with moderate or strong ribavirin alone (~ 36% vs 17%, respectively).
inhibitors or inducers of CYP3A may Leukopenia, thrombocytopenia, increased
significantly increase or decrease the plasma serum bilirubin levels, hyperuricemia, and
concentration of simeprevir. anorectal burning may also occur.
The presence of the NS3 Q80K
polymorphism at baseline is associated with
reduced efficacy of therapy, and screening is RIBAVIRIN
recommended prior to the initiation of
Ribavirin is a guanosine analog that is
therapy. Emergence of amino acid
phosphorylated intracellularly by host cell
substitutions resulting in decreased drug
susceptibility has been documented during enzymes.
therapy and may be associated with reduced Although its mechanism of action has not been
responsiveness. fully elucidated, it appears to interfere with the
Reported adverse events include synthesis of guanosine triphosphate, to inhibit
photosensitivity reaction and rash (most capping of viral messenger RNA, and to inhibit
common within the first 4 weeks of therapy). the viral RNA-dependent polymerase of certain
Since simeprevir contains a sulfa moiety, viruses.
caution should be used in patients with a Ribavirin triphosphate inhibits the replication of
history of sulfa allergy. a wide range of DNA and RNA viruses,
including influenza A and B, parainfluenza,
Telaprevir respiratory syncytial virus, paramyxoviruses,
Therapy with telaprevir plus peginterferonn HCV, and HIV-1.
and ribavirin is administered for at least 12 The absolute oral bioavailability of ribavirin is
weeks in treatment-naïve patients with HCV 45–64%, increases with high-fat meals, and
infection. decreases with co-administration of antacids.
As with boceprevir, the duration of therapy is Plasma protein binding is negligible, volume of
dependent on the achievement of undetectable distribution is large, and cerebrospinal fluid
virus. levels are about 70% of those in plasma.
Telaprevir must be taken with food to Ribavirin elimination is primarily through the
maximize absorption. urine; therefore, clearance is decreased in
It is 59–76% bound to plasma proteins and patients with creatinine clearances less than 30
the effective half-life at steady state is 9–11
mL/min.
hours.
Higher doses of ribavirin (ie, 1000–1200 mg/d,
Telaprevir is metabolized by the CYP
pathways in the liver and is an inhibitor of according to weight, rather than 800 mg/d) or a
CYP3A4 and P-glycoprotein. longer duration of therapy or both may be more
Co-administration of telaprevir with efficacious in those with a lower likelihood of
numerous drugs is contraindicated, including response to therapy (eg, those with genotype 1
rifampin, ergot derivatives, cisapride, or 4) or in those who have relapsed.
lovastatin, simvastatin, alfuzosin, sildenafil or This must be balanced with an increased
tadalafil when used for pulmonary likelihood of toxicity.
hypertension, pimozide, St. John’s wort, A dose-dependent hemolytic anemia occurs in
triazolam, and midazolam. 10–20% of patients.
The dosage of telaprevir must be increased Other potential adverse effects are depression,
when coadministered with efavirenz, due to
fatigue, irritability, rash, cough, insomnia,
lowered levels of telaprevir.
nausea, and pruritus. Contraindications to
The most commonly reported adverse effects
ribavirin therapy include anemia, end-stage renal form and have recently expanded their host
failure, ischemic vascular disease, and range to cause both avian and human disease.
pregnancy. Of particular concern is the avian H5N1 virus,
Ribavirin is teratogenic and embryotoxic in which first caused human infection (including
animals as well as mutagenic in mammalian severe disease and death) in 1997 and has
cells. become endemic in Southeast Asian poultry
Patients exposed to the drug should not conceive since 2003.
children for at least 6 months thereafter. To date, the spread of H5N1 virus from person
to person has been rare, limited, and
NEW & INVESTIGATIONAL AGENTS
unsustained.
Second-generation NS3/NS4A protease However, the emergence of the 2009 H1N1
inhibitors (eg, faldaprevir, simeprevir, influenza virus (previously called “swine flu”) in
asunaprevir), nucleoside/nucleotide NS5B 2009–2010 caused the first influenza pandemic
polymerase inhibitors (eg, sofosbuvir, see (ie, global outbreak of disease caused by a new
above), and non-nucleoside NS5B polymerase flu virus) in more than 40 years.
inhibitors (eg, deleobuvir) are currently under
OSELTAMIVIR & ZANAMIVIR
clinical investigation. The neuraminidase inhibitors oseltamivir and
The goal is to identify potent and well tolerated zanamivir, analogs of sialic acid, interfere
regimens that do not require concurrent with release of progeny influenza virus from
administration of interferon or ribavirin; in infected host cells, thus halting the spread of
addition agents are needed with activity against infection within the respiratory tract.
HCV genotypes other than 1 (such as These agents competitively and reversibly
sofosbuvir). interact with the active enzyme site to inhibit
Other classes of agents in development include viral neuraminidase activity at low nanomolar
NS5A inhibitors (eg, daclatasvir), p7 and NS4B concentrations.
inhibitors, cyclophilin inhibitors, and antisense Inhibition of viral neuraminidase results in
oligonucleotides inhibiting miR122 (eg, clumping of newly released influenza virions
to each other and to the membrane of the
miravirsen).
infected cell.
ANTI-INFLUENZA AGENTS Unlike amantadine and rimantadine,
oseltamivir and zanamivir have activity
Influenza virus strains are classified by their against both influenza A and influenza B
core proteins (ie, A, B, or C), species of origin viruses.
(eg, avian, swine), and geographic site of Early administration is crucial because
isolation. replication of influenza virus peaks at 24–72
Influenza A, the only strain that causes hours after the onset of illness.
pandemics, is classified into 16 H Initiation of a 5-day course of therapy within
(hemagglutinin) and 9 N (neuraminidase) known 48 hours after the onset of illness decreases
the duration of symptoms, viral shedding and
subtypes based on surface proteins.
transmission, and the rate of complications
Although influenza B viruses usually infect only
such as pneumonia, asthma, hospitalization,
people, influenza A viruses can infect a variety and mortality.
of animal hosts. Once-daily prophylaxis is 70–90% effective
Current influenza A subtypes that are circulating in preventing disease after exposure.
among worldwide populations include H1N1, Oseltamivir is an orally administered prodrug
H1N2, and H3N2. Fifteen subtypes are known that is activated by hepatic esterases and
to infect birds, providing an extensive reservoir. widely distributed throughout the body.
Although avian influenza subtypes are typically The dosage is 75 mg twice daily for 5 days
highly species-specific, they have on rare for treatment and 75 mg once daily for
occasions crossed the species barrier to infect prevention.
humans and cats. Oral bioavailability is approximately 80%,
Viruses of the H5 and H7 subtypes (eg, H5N1, plasma protein binding is low, and
concentrations in the middle ear and sinus
H7N7, and H7N3) may rapidly mutate within
fluid are similar to those in plasma.
poultry flocks from a low to high pathogenic
The half-life of oseltamivir is 6–10 hours, and
excretion is by glomerular filtration and emerge during treatment.
tubular secretion.
Probenecid reduces renal clearance of
oseltamivir by 50%. AMANTADINE & RIMANTADINE
Serum concentrations of oseltamivir Amantadine (1-aminoadamantane
carboxylate, the active metabolite of hydrochloride) and its α-methyl derivative,
oseltamivir, increase with declining renal rimantadine, are tricyclic amines of the
function; therefore, dosage should be adjusted adamantane family that block the M2 proton
in patients with renal insufficiency. ion channel of the virus particle and inhibit
Potential adverse effects include nausea, uncoating of the viral RNA within infected
vomiting, and headache. host cells, thus preventing its replication.
Taking oseltamivir with food does not They are active against influenza A only.
interfere with absorption and may decrease Rimantadine is four to ten times more active
nausea and vomiting. than amantadine in vitro.
Fatigue and diarrhea have also been reported Amantadine is well absorbed and 67%
and appear to be more common with protein-bound. Its plasma half-life is 12–18
prophylactic use. hours and varies by creatinine clearance.
Rash is rare. Rimantadine is about 40% protein-bound and
Neuropsychiatric events (self-injury or has a half-life of 24–36 hours.
delirium) have been reported, particularly in Nasal secretion and salivary levels
adolescents and adults living in Japan. approximate those in the serum, and
Zanamivir is administered directly to the cerebrospinal fluid levels are 52–96% of
respiratory tract via inhalation. those in the serum; nasal mucus
Ten to twenty percent of the active compound concentrations of rimantadine average 50%
reaches the lungs, and the remainder is higher than those in plasma.
deposited in the oropharynx. Amantadine is excreted unchanged in the
The concentration of the drug in the urine, whereas rimantadine undergoes
respiratory tract is estimated to be more than extensive metabolism by hydroxylation,
1000 times the 50% inhibitory concentration conjugation, and glucuronidation before
for neuraminidase, and the pulmonary half- urinary excretion.
life is 2.8 hours. Dose reductions are required for both agents
Five to fifteen percent of the total dose (10 in the elderly and in patients with renal
mg twice daily for 5 days for treatment and 10 insufficiency, and for rimantadine in patients
mg once daily for prevention) is absorbed and with marked hepatic insufficiency.
excreted in the urine with minimal In the absence of resistance, both amantadine
metabolism. and rimantadine, at 100 mg twice daily or 200
Potential adverse effects include cough, mg once daily, are 70–90% protective in the
bronchospasm (occasionally severe), prevention of clinical illness when initiated
reversible decrease in pulmonary function, before exposure.
and transient nasal and throat discomfort. When begun within 1–2 days after the onset
Zanamivir administration is not recommended of illness, the duration of fever and systemic
for patients with underlying airway disease. symptoms is reduced by 1–2 days.
Both oseltamivir and zanamivir are available However, due to high rates of resistance in
in intravenous formulations on a both H1N1 and H3N2 viruses, these agents
compassionate use basis from the are no longer recommended for the prevention
manufacturer. or treatment of influenza.
Although resistance to oseltamivir and The most common adverse effects are
zanamivir may emerge during therapy and be gastrointestinal (nausea, anorexia) and central
transmissible, nearly 100% of strains of nervous system (nervousness, difficulty in
H1N1, H3N2, and influenza B virus tested by concentrating, insomnia, light-headedness).
the Centers for Diseases Control for the 2012- More serious side effects (eg, marked
2013 season retained susceptibility to both behavioral changes, delirium, hallucinations,
agents. agitation, and seizures) may be due to
Oseltamivir resistance, however, has been alteration of dopamine neurotransmission (see
documented in strains of the novel avian Chapter 28); are less frequent with
H7N9 virus, in one instance appearing to rimantadine than with amantadine; are
associated with high plasma concentrations; gained widespread use.
may occur more frequently in patients with Systemic absorption is low (< 1%).
renal insufficiency, seizure disorders, or Aerosolized ribavirin may cause conjunctival
advanced age; and may increase with or bronchial irritation and the aerosolized
concomitant antihistamines, anticholinergic drug may precipitate on contact lenses.
drugs, hydrochlorothiazide, and trimethoprim- Ribavirin is teratogenic and embryotoxic.
sulfamethoxazole. Health care workers and pregnant women
Clinical manifestations of anticholinergic should be protected against extended
activity tend to be present in acute amantadine inhalation exposure.
overdose. Intravenous ribavirin decreases mortality in
Both agents are teratogenic and embryotoxic patients with Lassa fever and other viral
in rodents, and birth defects have been hemorrhagic fevers if started early.
reported after exposure during pregnancy. High concentrations inhibit West Nile virus in
vitro, but clinical data are lacking.
Clinical benefit has been reported in cases of
INVESTIGATIONAL AGENTS severe measles pneumonitis and certain
encephalitides, and continuous infusion of
The neuraminidase inhibitor peramivir, a
ribavirin has decreased virus shedding in
cyclopentane analog, has activity against both
several patients with severe lower respiratory
influenza A and B viruses. tract influenza or parainfluenza infections.
Peramivir received temporary emergency use At steady state, cerebrospinal fluid levels are
authorization by FDA for intravenous about 70% of those in plasma.
administration in November 2009 due to the
H1N1 pandemic, but is not now approved for
use in the USA. PALIVIZUMAB
Reported side effects include diarrhea, nausea, Palivizumab is a humanized monoclonal
vomiting, and neutropenia. antibody directed against an epitope in the A
antigen site on the F surface protein of RSV.
A long-acting neuraminidase inhibitor,
It is licensed for the prevention of RSV
laninamivir octanoate, may retain activity
infection in high-risk infants and children,
against oseltamivir-resistant virus. such as premature infants and those with
DAS181 is a host-directed antiviral agent that bronchopulmonary dysplasia or congenital
acts by removing the virus receptor, sialic acid, heart disease.
from adjacent glycan structures. A placebo-controlled trial using once-monthly
intramuscular injections (15 mg/kg) for 5
OTHER ANTIVIRAL AGENTS months beginning at the start of the RSV
season demonstrated a 55% reduction in the
INTERFERONS risk of hospitalization for RSV in treated
Interferons have been studied for numerous patients, as well as decreases in the need for
clinical indications. In addition to HBV and supplemental oxygen, the illness severity
HCV infections (see Antihepatitis Agents), score, and the need for intensive care.
intralesional injection of interferon alfa-2b or Although resistant strains have been isolated
alfa-n3 may be used for treatment of in the laboratory, no resistant clinical isolates
condylomata acuminata (see Chapter 61). have yet been identified.
Potential adverse effects include upper
respiratory tract infection, fever, rhinitis, rash,
RIBAVIRIN diarrhea, vomiting, cough, otitis media, and
In addition to oral administration for HCV elevation in serum aminotransferase levels.
infection in combination with interferon alfa Agents under investigation for the treatment
(see Antihepatitis Agents), aerosolized or prophylaxis of patients with RSV infection
ribavirin is administered by nebulizer (20 include the RNA interference (RNAi)
mg/mL for 12–18 hours per day) to children therapeutic ALN-RSV01and the
and infants with severe respiratory syncytial benzodiazepine RSV604.
virus (RSV) bronchiolitis or pneumonia to
reduce the severity and duration of illness.
Aerosolized ribavirin has also been used to IMIQUIMOD
treat influenza A and B infections but has not Imiquimod is an immune response modifier
shown to be effective in the topical treatment
of external genital and perianal warts (ie,
condyloma acuminatum; see Chapter 61).
The 5% cream is applied three times weekly
and washed off 6–10 hours after each
application. Recurrences appear to be less
common than after ablative therapies.
Imiquimod may also be effective against
molluscum contagiosum but is not licensed in
the United States for this indication.
Local skin reactions are the most common
adverse effect; these tend to resolve within
weeks after therapy.
However, pigmentary skin changes may
persist.
Systemic adverse effects such as fatigue and
influenza-like syndrome have occasionally
been reported.

You might also like