You are on page 1of 41

Antiviral Agents of time (interferon alfa/ribavirin for HCV),

whereas still others require multiple drug


Viruses are obligate intracellular parasites; therapy for indefinite periods (HIV).
their replication depends primarily on In chronic illnesses such as viral hepatitis and
synthetic processes of the host cell. HIV infection, potent inhibition of viral
Therefore, to be effective, antiviral agents replication is crucial in limiting the extent of
must either block viral entry into or exit from systemic damage.
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
potential targets for antiviral therapy.
Recent research has focused on identifying
agents with greater selectivity, higher Viral replication requires several steps (Figure
potency, in vivo stability, and reduced 49–1):
toxicity. (1) attachment of the virus to receptors on
Antiviral therapy is now available for the host cell surface;
herpesviruses, hepatitis C virus (HCV), (2) entry of the virus through the host cell
hepatitis B virus (HBV), papillomavirus, membrane;
influenza, human immunodeficiency virus (3) uncoating of viral nucleic acid;
(HIV), and respiratory syncytial virus (RSV). (4) synthesis of early regulatory proteins, eg,
Antiviral drugs share the common property nucleic acid polymerases;
of being virustatic; they are active only (5) synthesis of new viral RNA or DNA;
against replicating viruses and do not affect (6) integration into the nuclear genome;
latent virus. (7) synthesis of late, structural proteins;
Whereas some infections require (8) assembly (maturation) of viral particles;
monotherapy for brief periods of time (eg, and (9) release from the cell.
acyclovir for herpes simplex virus), others Antiviral agents can potentially target any of
require dual therapy for prolonged periods these steps.
Comparative trials have demonstrated
similar efficacies of these three agents for the
treatment of HSV but modest superiority of
famciclovir 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
AGENTS TO TREAT HERPES SIMPLEX VIRUS DNA template as an irreversible complex;
(HSV) & VARICELLA-ZOSTER VIRUS (VZV) and chain termination following
INFECTIONS incorporation into the viral DNA.
The bioavailability of oral acyclovir is low
(15–20%) and is unaffected by food.
Three oral nucleoside analogs are licensed
An intravenous formulation is available.
for the treatment of HSV and VZV infections:
Topical formulations produce high
acyclovir, valacyclovir, and famciclovir.
concentrations in herpetic lesions, but
They have similar mechanisms of action and systemic concentrations are undetectable
comparable indications for clinical use; all are by this route.
well tolerated. Acyclovir is cleared primarily by glomerular
Acyclovir has been the most extensively filtration and tubular secretion.
studied; it was licensed first and is the only The half-life is 2.5–3 hours in patients with
one of the three that is available for normal renal function and 20 hours in
intravenous use in the United States. patients with anuria.
Acyclovir diffuses readily into most tissues neonatal HSV infection, and serious HSV or
and body fluids. VZV infections (Table 49–1).
Cerebrospinal fluid concentrations are 20– In neonates with central nervous system
50% of serum values. HSV, oral acyclovir suppression for 6
Oral acyclovir has multiple uses. In first months following acute treatment
episodes of genital herpes, oral acyclovir improves neurodevelopmental outcomes.
shortens the duration of symptoms by In immunocompromised patients with
approximately 2 days, the time to lesion VZV infection, intravenous acyclovir
healing by 4 days, and the duration of viral reduces the incidence of cutaneous and
shedding by 7 days. visceral dissemination.
In recurrent anogenital herpes, the time Topical acyclovir cream is substantially less
course is shortened by 1–2 days. effective than oral therapy for primary HSV
Treatment of first-episode genital herpes infection.
does not alter the frequency or severity of It is of no benefit in treating recurrent
recurrent outbreaks. genital herpes.
Long-term suppression with oral acyclovir Resistance to acyclovir can develop in HSV
in patients with frequent recurrences of or VZV through alteration in either the viral
genital herpes decreases the frequency of thymidine kinase or the DNA polymerase,
symptomatic recurrences and of and clinically resistant infections have
asymptomatic viral shedding, thus been reported in immunocompromised
decreasing the rate of sexual transmission. hosts.
However, outbreaks may resume upon Most clinical isolates are resistant on the
discontinuation of suppressive acyclovir. basis of deficient thymidine kinase activity
Oral acyclovir is only modestly beneficial in and thus are cross-resistant to valacyclovir,
recurrent herpes labialis. famciclovir, and ganciclovir.
In contrast, acyclovir therapy significantly Agents such as foscarnet, cidofovir, and
decreases the total number of lesions, trifluridine do not require activation by
duration of symptoms, and viral shedding viral thymidine kinase and thus have
in patients with varicella (if begun within preserved activity against the most
24 hours after the onset of rash) or prevalent acyclovir-resistant strains (Figure
cutaneous zoster (if begun within 72 49–2).
hours); the risk of post-herpetic neuralgia Acyclovir is generally well tolerated,
is also reduced if treatment is initiated although nausea, diarrhea, and headache
early. may occur.
However, because VZV is less susceptible Intravenous infusion may be associated
to acyclovir than HSV, higher doses are with reversible renal toxicity (ie, crystalline
required (Table 49–1). nephropathy or interstitial nephritis) or
When given prophylactically to patients neurologic effects (eg, tremors, delirium,
undergoing organ transplantation, oral or seizures).
intravenous acyclovir prevents reactivation However, these are uncommon with
of HSV and VZV infection. adequate hydration and avoidance of
Evidence from clinical trials suggests that rapid infusion rates.
the use of daily acyclovir (400 mg twice High doses of acyclovir cause
daily) may reduce the plasma viral load of chromosomal damage and testicular
HIV-1 and the risk of HIV-associated atrophy in rats, but there has been no
disease progression in individuals dually evidence of teratogenicity, reduction in
infected with HSV-2 and HIV-1. sperm production, or cytogenetic
Intravenous acyclovir is the treatment of alterations in peripheral blood
choice for herpes simplex encephalitis, lymphocytes in patients receiving daily
suppression of genital herpes for more oral acyclovir and approximate those
than 10 years. achieved with intravenous acyclovir.
A recent study found no evidence of Oral bioavailability is 54–70%, and
increased birth defects in 1150 infants who cerebrospinal fluid levels are about 50% of
were exposed to acyclovir during the first those in serum.
trimester. Elimination half-life is 2.5–3.3 hours.
In fact, the American College of Twice-daily valacyclovir is effective for
Obstetricians and Gynecologists treatment of first or recurrent genital
recommends suppressive acyclovir herpes and varicella and zoster infections;
therapy beginning at week 36 in pregnant it is approved for use as a 1-day treatment
women with active recurrent genital for orolabial herpes and as suppression of
herpes to reduce the risk of recurrence at frequently recurring genital herpes (Table
delivery and possibly the need for 49–1).
cesarean section. Once-daily dosing of valacyclovir for
The impact of this intervention on chronic suppression in persons with
neonatal infection has not been recurrent genital herpes has been shown
established. to markedly decrease the risk of sexual
Concurrent use of nephrotoxic agents may transmission.
enhance the potential for nephrotoxicity. In comparative trials with acyclovir for the
Probenecid and cimetidine decrease treatment of patients with zoster, rates of
acyclovir clearance and increase exposure. cutaneous healing were similar, but
Somnolence and lethargy may occur in valacyclovir was associated with a shorter
patients receiving concomitant zidovudine duration of zoster-associated pain.
and acyclovir. 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
VALACYCLOVIR
evidence of increased birth defects in 181
Valacyclovir is the L-valyl ester of acyclovir.
infants who were exposed to valacyclovir
It is rapidly converted to acyclovir after oral
during the first trimester.
administration via first-pass enzymatic
hydrolysis in the liver and intestine,
resulting in serum levels that are three to
five times greater than those achieved with
FAMCICLOVIR Oral famciclovir is generally well tolerated,
Famciclovir is the diacetyl ester prodrug of although headache, nausea, or diarrhea
6-deoxypenciclovir, an acyclic guanosine may occur.
analog (eFigure 49–2.1). As with acyclovir, testicular toxicity has
After oral administration, famciclovir is been demonstrated in animals receiving
rapidly deacetylated and oxidized by first- repeated doses.
pass metabolism to penciclovir. However, men receiving daily famciclovir
It is active in vitro against HSV-1, HSV-2, (250 mg every 12 hours) for 18 weeks had
VZV, EBV, and HBV. no changes in sperm morphology or
As with acyclovir, activation by motility.
phosphorylation is catalyzed by the virus- In a recent study, there was no evidence of
specified thymidine kinase in infected cells, increased birth defects in 32 infants who
followed by competitive inhibition of the were exposed to famciclovir during the
viral DNA polymerase to block DNA first trimester.
synthesis. The incidence of mammary
Unlike acyclovir, however, penciclovir does adenocarcinoma was increased in female
not cause chain termination. rats receiving famciclovir for 2 years.
Penciclovir triphosphate has lower affinity
for the viral DNA polymerase than
acyclovir triphosphate, but it achieves PENCICLOVIR
higher intracellular concentrations. The guanosine analog penciclovir, the
The most commonly encountered clinical active metabolite of famciclovir, is
mutants of HSV are thymidine kinase- available for topical use.
deficient; these are cross-resistant to Penciclovir cream (1%) shortened the
acyclovir and famciclovir. median duration of recurrent herpes
The bioavailability of penciclovir from labialis by ~ 17 hours compared to
orally administered famciclovir is 70%. placebo when applied within 1 hour of the
The intracellular half-life of penciclovir onset of prodromal symptoms and
triphosphate is prolonged, at 7–20 hours. continued every 2 hours during waking
Penciclovir is excreted primarily in the hours for 4 days.
urine. Adverse effects are uncommon, other than
Oral famciclovir is effective for the application site reactions in ~1%.
treatment of first and recurrent genital
herpes, for chronic daily suppression of
DOCOSANOL
genital herpes, for treatment of herpes
Docosanol is a saturated 22-carbon
labialis, and for the treatment of acute
aliphatic alcohol that inhibits fusion
zoster (Table 49–1).
between the host cell plasma membrane
One-day usage of famciclovir significantly
and the HSV envelope, thereby preventing
accelerates time to healing of recurrent
viral entry into cells and subsequent viral
genital herpes and of herpes labialis.
replication.
Comparison of famciclovir to valacyclovir
Topical docosanol 10% cream is available
for treatment of herpes zoster in
without a prescription.
immunocompetent patients showed
When applied within 12 hours of the onset
similar rates of cutaneous healing and pain
of prodromal symptoms, five times daily,
resolution; both agents shortened the
median healing time was shortened by 18
duration of zoster-associated pain
hours compared with placebo in recurrent
compared with acyclovir.
orolabial herpes.
Application site reactions occur in ~2%.
The availability of oral valganciclovir has
decreased the use of intravenous ganciclovir,
TRIFLURIDINE
intravenous foscarnet, and intravenous
Trifluridine (trifluorothymidine) is a
cidofovir for the prophylaxis and treatment
fluorinated pyrimidine nucleoside that
inhibits viral DNA synthesis in HSV-1, HSV- of end-organ CMV disease (Table 49–2).
2, CMV, vaccinia, and some adenoviruses. Oral valganciclovir has replaced oral
It is phosphorylated intracellularly by host ganciclovir because of its lower pill burden.
cell enzymes, and then competes with
thymidine triphosphate for incorporation
by the viral DNA polymerase (Figure 49–2).
Incorporation of trifluridine triphosphate
into both viral and host DNA prevents its
systemic use.
Application of a 1% solution is effective in
treating keratoconjunctivitis and recurrent GANCICLOVIR
epithelial keratitis due to HSV-1 or HSV-2. Ganciclovir is an acyclic guanosine analog
Cutaneous application of trifluridine (eFigure 49–2.1) that requires activation by
solution, alone or in combination with triphosphorylation before inhibiting the
interferon alfa, has been used successfully viral DNA polymerase.
in the treatment of acyclovir-resistant HSV Initial phosphorylation is catalyzed by the
infections. virus-specified protein kinase
phosphotransferase UL97 in CMV-infected
cells.
INVESTIGATIONAL AGENTS
The activated compound competitively
Valomaciclovir is an inhibitor of the viral
inhibits viral DNA polymerase and causes
DNA polymerase; it is currently under
termination of viral DNA elongation
clinical evaluation for the treatment of
(Figure 49–2).
patients with acute zoster and acute EBV
Ganciclovir has in vitro activity against
infection (infectious mononucleosis).
CMV, HSV, VZV, EBV, HHV-6, and HHV-8.
Its activity against CMV is up to 100 times
AGENTS TO TREAT CYTOMEGALOVIRUS (CMV) greater than that of acyclovir.
INFECTIONS Ganciclovir is administered intravenously;
the bioavailability of oral ganciclovir is
poor, and it is no longer available in the US.
CMV infections occur primarily in the setting
Ganciclovir gel is available for the
of advanced immunosuppression and are
treatment of acute herpetic keratitis.
typically due to reactivation of latent
Cerebrospinal fluid concentrations are
infection. approximately 50% of serum
Dissemination of infection results in end- concentrations.
organ disease, including retinitis, colitis, The elimination half-life is 4 hours, and the
esophagitis, central nervous system disease, intracellular half-life is prolonged at 16–24
and pneumonitis. hours.
Although the incidence in HIV-infected Clearance of the drug is linearly related to
patients has markedly decreased with the creatinine clearance.
advent of potent anti-retroviral therapy, Ganciclovir is readily cleared by
clinical reactivation of CMV infection after hemodialysis.
organ transplantation is still prevalent.
Intravenous ganciclovir has been shown to Myelosuppression may be additive in
delay progression of CMV retinitis in patients receiving concurrent zidovudine,
immunocompromised patients. azathioprine, or mycophenolate mofetil.
Dual therapy with foscarnet and Other potential adverse effects are nausea,
ganciclovir is more effective in delaying diarrhea, fever, rash, headache, insomnia,
progression of retinitis than either drug and peripheral neuropathy.
alone in patients with AIDS (see Foscarnet), Central nervous system toxicity (confusion,
although adverse effects are compounded. seizures, psychiatric disturbance) and
Intravenous ganciclovir is also used to hepatotoxicity have been rarely reported.
treat CMV colitis, esophagitis, and Intravitreal ganciclovir has been associated
pneumonitis (the latter often in with vitreous hemorrhage and retinal
combination with intravenous detachment.
cytomegalovirus immunoglobulin) in Ganciclovir is mutagenic in mammalian
immunocompromised patients. cells and carcinogenic and embryotoxic at
Intravenous ganciclovir, followed by either high doses in animals and causes
oral ganciclovir or high-dose oral acyclovir, aspermatogenesis; the clinical significance
reduced the risk of CMV infection in of these preclinical data is unclear.
transplant recipients. Levels of ganciclovir may rise in patients
Limited data in infants with symptomatic concurrently taking probenecid or
congenital neurologic CMV disease trimethoprim.
suggest that treatment with IV ganciclovir Concurrent use of ganciclovir with
may reduce hearing loss. didanosine may result in increased levels
The risk of Kaposi’s sarcoma is reduced in of didanosine.
AIDS patients receiving long-term
ganciclovir, presumably because of activity
against HHV-8. VALGANCICLOVIR
Intravitreal injections of ganciclovir may be Valganciclovir is an L-valyl ester prodrug of
used to treat CMV retinitis. ganciclovir that exists as a mixture of two
Concurrent therapy with a systemic anti- diastereomers.
CMV agent is necessary to prevent other After oral administration, both
sites of end-organ CMV disease. diastereomers are rapidly hydrolyzed to
The intraocular ganciclovir implant is no ganciclovir by esterases in the intestinal
longer available in the USA. Resistance to wall and liver.
ganciclovir increases with duration of use. Valganciclovir is well absorbed; the
The more common mutation, in UL97, bioavailability of oral valganciclovir is 60%
results in decreased levels of the and it is recommended that the drug be
triphosphorylated (ie, active) form of taken with food.
ganciclovir. The AUC0–24h resulting from
The less common UL54 mutation in DNA valganciclovir (900 mg once daily) is similar
polymerase results in higher levels of to that after 5 mg/kg once daily of
resistance and potential cross-resistance intravenous ganciclovir and approximately
with cidofovir and foscarnet. 1.65 times that of oral ganciclovir.
Antiviral susceptibility testing is The major route of elimination is renal,
recommended in patients in whom through glomerular filtration and active
resistance is suspected clinically. tubular secretion.
The most common adverse effect of Plasma concentrations of valganciclovir
intravenous ganciclovir treatment is are reduced approximately 50% by
myelosuppression, which although hemodialysis.
reversible may be dose-limiting.
Valganciclovir is as effective as intravenous The dosage of foscarnet must be titrated
ganciclovir for the treatment of CMV according to the patient’s calculated
retinitis and is also indicated for the creatinine clearance before each infusion.
prevention of CMV disease in high-risk Use of an infusion pump to control the rate
solid organ and bone marrow transplant of infusion is important to prevent toxicity,
recipients. and large volumes of fluid are required
Adverse effects, drug interactions, and because of the drug’s poor solubility.
resistance patterns are the same as those The combination of ganciclovir and
associated with ganciclovir. foscarnet is synergistic in vitro against
CMV and has been shown to be superior
to either agent alone in delaying
FOSCARNET progression of retinitis; however, toxicity is
Foscarnet (phosphonoformic acid) is an also increased when these agents are
inorganic pyrophosphate analog that administered concurrently.
inhibits herpesvirus DNA polymerase, RNA As with ganciclovir, a decrease in the
polymerase, and HIV reverse transcriptase incidence of Kaposi’s sarcoma has been
directly without requiring activation by observed in patients who have received
phosphorylation. long-term foscarnet.
Foscarnet blocks the pyrophosphate Foscarnet has been administered
binding site of these enzymes and inhibits intravitreally for the treatment of CMV
cleavage of pyrophosphate from retinitis in patients with AIDS, but data
deoxynucleotide triphosphates. regarding efficacy and safety are
It has in vitro activity against HSV, VZV, incomplete.
CMV, EBV, HHV-6, HHV-8, HIV-1, and HIV- Resistance to foscarnet in HSV and CMV
2. isolates is due to point mutations in the
Foscarnet is available in an intravenous DNA polymerase gene and is typically
formulation only; poor oral bioavailability associated with prolonged or repeated
and gastrointestinal intolerance preclude exposure to the drug.
oral use. Mutations in the HIV-1 reverse
Cerebrospinal fluid concentrations are 43– transcriptase gene have also been
67% of steady-state serum concentrations. described.
Although the mean plasma half-life is 3–7 Although foscarnet-resistant CMV isolates
hours, up to 30% of foscarnet may be are typically cross-resistant to ganciclovir,
deposited in bone, with a half-life of foscarnet activity is usually maintained
several months. against ganciclovir- and cidofovir-resistant
The clinical repercussions of this are isolates of CMV.
unknown. Potential adverse effects of foscarnet
Clearance of foscarnet is primarily renal include renal impairment, hypo- or
and is directly proportional to creatinine hypercalcemia, hypo- or
clearance. hyperphosphatemia, hypokalemia, and
Serum drug concentrations are reduced hypomagnesemia.
approximately 50% by hemodialysis. Saline preloading helps prevent
Foscarnet is effective in the treatment of nephrotoxicity, as does avoidance of
end-organ CMV disease (ie, retinitis, colitis, concomitant administration of drugs with
and esophagitis), including ganciclovir- nephrotoxic potential (eg, amphotericin B,
resistant disease; it is also effective against pentamidine, aminoglycosides).
acyclovir-resistant HSV and VZV infections. The risk of severe hypocalcemia, caused by
chelation of divalent cations, is increased
with concomitant use of pentamidine.
Genital ulcerations associated with Elimination is by active renal tubular
foscarnet therapy may be due to high secretion.
levels of ionized drug in the urine. High-flux hemodialysis reduces serum
Nausea, vomiting, anemia, elevation of levels of cidofovir by approximately 75%.
liver enzymes, and fatigue have been Intravenous cidofovir is effective for the
reported; the risk of anemia may be treatment of CMV retinitis and is used
additive in patients receiving concurrent experimentally to treat adenovirus, human
zidovudine. papillomavirus, BK polyomavirus, vaccinia,
Central nervous system toxicity includes and poxvirus infections.
headache, hallucinations, and seizures; the Intravenous cidofovir must be
risk of seizures may be increased with administered with high-dose probenecid
concurrent use of imipenem. (2 g at 3 hours before the infusion and 1 g
Foscarnet caused chromosomal damage in at 2 and 8 hours after), which blocks active
preclinical studies. tubular secretion and decreases
nephrotoxicity.
Before each infusion, cidofovir dosage
CIDOFOVIR must be adjusted for alterations in the
Cidofovir (eFigure 49–2.1) is a cytosine calculated creatinine clearance or for the
nucleotide analog with in vitro activity presence of urine protein, and aggressive
against CMV, HSV-1, HSV-2, VZV, EBV, adjunctive hydration is required.
HHV-6, HHV-8, adenovirus, poxviruses, Initiation of cidofovir therapy is
polyomaviruses, and human contraindicated in patients with existing
papillomavirus. renal insufficiency.
In contrast to ganciclovir, phosphorylation Direct intravitreal administration of
of cidofovir to the active diphosphate is cidofovir is not recommended because of
independent of viral enzymes (Figure 49– ocular toxicity.
2); thus activity is maintained against The primary adverse effect of intravenous
thymidine kinase-deficient or -altered cidofovir is a dose-dependent proximal
strains of CMV or HSV. tubular nephrotoxicity, which may be
Cidofovir diphosphate acts both as a reduced with prehydration using normal
potent inhibitor of and as an alternative saline.
substrate for viral DNA polymerase, Proteinuria, azotemia, metabolic acidosis,
competitively inhibiting DNA synthesis and Fanconi’s syndrome may occur.
and becoming incorporated into the viral Concurrent administration of other
DNA chain. potentially nephrotoxic agents (eg,
Cidofovir-resistant isolates tend to be amphotericin B, aminoglycosides,
cross-resistant with ganciclovir but retain nonsteroidal anti-inflammatory drugs,
susceptibility to foscarnet. pentamidine, foscarnet) should be
Although the terminal half-life of cidofovir avoided.
is approximately 2.6 hours, the active Prior administration of foscarnet may
metabolite cidofovir diphosphate has a increase the risk of nephrotoxicity.
prolonged intracellular half-life of 17–65 Other potential adverse effects include
hours, thus allowing infrequent dosing. uveitis, ocular hypotony, and neutropenia
A separate metabolite, cidofovir (15– 24%).
phosphocholine, has a half-life of at least Concurrent probenecid use may result in
87 hours and may serve as an intracellular other toxicities or drug-drug interactions
reservoir of active drug. (see Chapter 36).
Cerebrospinal fluid penetration is poor.
Cidofovir is mutagenic, gonadotoxic, and for any given patient are tolerability,
embryotoxic, and causes hypospermia and convenience, and optimization of adherence.
mammary adenocarcinomas in animals. As new agents have become available,
several older ones have had diminished
usage, because of either suboptimal safety or
ANTIRETROVIRAL AGENTS
inferior antiviral efficacy.
Substantial advances have been made in Zalcitabine (ddC; dideoxycytidine), for
antiretroviral therapy since the introduction example, is no longer marketed.
of the first agent, zidovudine, in 1987. Decrease of the circulating viral load by
Six classes of antiretroviral agents are antiretroviral therapy is correlated with
currently available for use: enhanced survival as well as decreased
nucleoside/nucleotide reverse morbidity.
transcriptase inhibitors (NRTIs), non- Also, recent evidence suggests that in
nucleoside reverse transcriptase inhibitors addition to providing clinical benefits for the
(NNRTIs), protease inhibitors (PIs), fusion patient, the use of antiretroviral therapy
inhibitors, CCR5 co-receptor antagonists strongly reduces the risk for heterosexual HIV
(also called entry inhibitors), and HIV transmission.
integrase strand transfer inhibitors Discussion of antiretroviral agents in this
(INSTIs) (Table 49–3). chapter is specific to HIV-1.
These agents inhibit HIV replication at Patterns of susceptibility of HIV-2 to these
different parts of the cycle (Figure 49–3). agents may vary; however, there is generally
Structures of some of these drugs are shown innate resistance to the NNRTIs and lower
in eFigure 49–3.1. barriers of resistance to NRTIs and PIs; data
Greater knowledge of viral dynamics through regarding maraviroc are inconclusive.
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
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, stavudine plus
didanosine).
The NRTIs act by competitive inhibition of
HIV-1 reverse transcriptase; incorporation
into the growing viral DNA chain causes
premature chain termination due to
inhibition of binding with the incoming
nucleotide (Figure 49–3).
Each agent requires intracytoplasmic
activation via phosphorylation by cellular
enzymes to the triphosphate form.
Typical resistance mutations include M184V,
L74V, D67N, and M41L. Lamivudine or
emtricitabine therapy tends to select rapidly Since the drug is metabolized by alcohol
for the M184V mutation in regimens that are dehydrogenase, serum levels of abacavir
not fully suppressive. may be increased with concurrent alcohol
While the M184V mutation confers reduced (ie, ethanol) ingestion.
susceptibility to abacavir, didanosine, and Cerebrospinal fluid levels are
zalcitabine, its presence may restore approximately one-third those of plasma.
Abacavir is available in a fixed dose
phenotypic susceptibility to zidovudine.
formulation with lamivudine and also with
The K65R/N mutation is associated with
zidovudine plus lamivudine.
reduced susceptibility to tenofovir, abacavir,
High-level resistance to abacavir appears
lamivudine, and emtricitabine. to require at least two or three
All NRTIs may be associated with concomitant mutations and thus tends to
mitochondrial toxicity, probably owing to develop slowly.
inhibition of mitochondrial DNA polymerase Hypersensitivity reactions, occasionally
gamma. fatal, have been reported in up to 8% of
Less commonly, lactic acidosis with hepatic patients receiving abacavir and may be
steatosis may occur, which can be fatal. more severe in association with once-daily
NRTI treatment should be suspended in the dosing.
setting of rapidly rising aminotransferase Symptoms, which generally occur within
the first 6 weeks of therapy, include fever,
levels, progressive hepatomegaly, or
fatigue, nausea, vomiting, diarrhea, and
metabolic acidosis of unknown cause.
abdominal pain.
The thymidine analogs zidovudine and
Respiratory symptoms such as dyspnea,
stavudine may be particularly associated with pharyngitis, and cough may also be
dyslipidemia and insulin resistance. present, and skin rash occurs in about 50%
Also, some evidence suggests an increased of patients.
risk of myocardial infarction in patients The laboratory abnormalities of a mildly
receiving abacavir; this remains unproven. elevated serum aminotransferase or
creatine kinase level may be present but
are nonspecific.
Although the syndrome tends to resolve
quickly with discontinuation of
medication, rechallenge with abacavir
results in return of symptoms within hours
and may be fatal.
Screening for HLA-B*5701 before initiation
of abacavir therapy is recommended to
identify patients with a markedly increased
risk for abacavir-associated
hypersensitivity reaction.
Although the positive predictive value of
this test is only about 50%, it has a
ABACAVIR
negative predictive value approaching
Abacavir is a guanosine analog that is well
100%.
absorbed following oral administration
Other potential adverse events are rash,
(83%) and is unaffected by food.
fever, nausea, vomiting, diarrhea,
The serum half-life is 1.5 hours. The drug
headache, dyspnea, fatigue, and
undergoes hepatic glucuronidation and
pancreatitis (rare).
carboxylation.
In some studies but not in others, abacavir Due to an increased risk of lactic acidosis
has been associated with a higher risk of and hepatic steatosis when combined with
myocardial infarction. stavudine, this combination should be
Since abacavir may lower methadone avoided, especially during pregnancy.
levels, patients receiving these two agents Previously asymptomatic hyperuricemia
concurrently should be monitored for may precipitate attacks of gout in
signs of opioid withdrawal and may susceptible individuals; concurrent use of
require an increased dose of methadone. allopurinol may increase levels of
didanosine.
Reports of retinal changes and optic
DIDANOSINE neuritis in patients receiving didanosine,
Didanosine (ddI) is a synthetic analog of particularly in adults receiving high doses
deoxyadenosine. and in children, mandate periodic retinal
Oral bioavailability is approximately 40%. examinations.
Dosing on an empty stomach is optimal, Lipoatrophy appears to be more common
but buffered formulations are necessary to in patients receiving didanosine or other
prevent inactivation by gastric acid (Table thymidine analogs.
49–3). The buffer in didanosine tablets interferes
Cerebrospinal fluid concentrations of the with absorption of indinavir, delavirdine,
drug are approximately 20% of serum atazanavir, dapsone, itraconazole, and
concentrations. fluoroquinolone agents; therefore,
Serum half-life is 1.5 hours, but the administration should be separated in
intracellular half-life of the activated time.
compound is as long as 20–24 hours. Serum levels of didanosine are increased
The drug is eliminated by both cellular when co-administered with tenofovir or
metabolism and renal excretion. ganciclovir, and are decreased by
The major clinical toxicity associated with atazanavir, delavirdine, ritonavir,
didanosine therapy is dose-dependent tipranavir, and methadone (Table 49–4).
pancreatitis. Didanosine should not be used in
Other risk factors for pancreatitis (eg, combination with ribavirin.
alcohol abuse, hypertriglyceridemia) are
relative contraindications, and concurrent
use of drugs with the potential to cause EMTRICITABINE
pancreatitis, including zalcitabine, Emtricitabine (FTC) is a fluorinated analog
stavudine, ribavirin, and hydroxyurea, of lamivudine with a long intracellular half-
should be avoided (Table 49– 3). life (> 24 hours), allowing for once-daily
The risk of peripheral distal sensory dosing.
neuropathy, another potential toxicity, Oral bioavailability of the capsules is 93%
may be increased with concurrent use of and is unaffected by food, but penetration
stavudine, isoniazid, vincristine, or into the cerebrospinal fluid is low.
ribavirin. Elimination is by both glomerular filtration
Other reported adverse effects include and active tubular secretion.
diarrhea (particularly with the buffered The serum half-life is about 10 hours.
formulation), hepatitis, esophageal The oral solution, which contains
ulceration, cardiomyopathy, central propylene glycol, is contraindicated in
nervous system toxicity (headache, young children, pregnant women, patients
irritability, insomnia), and with renal or hepatic failure, and those
hypertriglyceridemia. using metronidazole or disulfiram.
Also, because of its activity against HBV, infected with HIV and HBV may be
patients co-infected with HIV and HBV associated with a flareof hepatitis.
should be closely monitored if treatment Lamivudine therapy rapidly selects for the
with emtricitabine is interrupted or M184V mutation in regimens that are not
discontinued, owing to the likelihood of fully suppressive.
hepatitis flare. Oral bioavailability exceeds 80% and is not
Emtricitabine is available in a fixed-dose food-dependent. In children, the average
formulation with tenofovir, either alone or cerebrospinal fluid:plasma ratio of
in combination with efavirenz, rilpivirine, lamivudine was 0.2.
or elvitegravir plus cobicistat (a boosting Serum half-life is 2.5 hours, whereas the
agent). intracellular half-life of the
Based on results of clinical trials, the triphosphorylated compound is 11–14
combination of tenofovir and hours.
emtricitabine is now recommended as pre- Most of the drug is eliminated unchanged
exposure prophylaxis to reduce HIV in the urine.
acquisition in men who have sex with men, Lamivudine remains one of the
in heterosexually active men and women, recommended antiretroviral agents in
and in injection drug users. pregnant women (Table 49–5).
Like lamivudine, the M184V/I mutation is Lamivudine is available in a fixed-dose
most frequently associated with formulation with zidovudine and also with
emtricitabine use and may emerge rapidly abacavir.
in patients receiving regimens that are not Potential adverse effects are headache,
fully suppressive. dizziness, insomnia, fatigue, dry mouth,
Because of their similar mechanisms of and gastrointestinal discomfort, although
action and resistance profiles, the these are typically mild and infrequent.
combination of lamivudine and Lamivudine’s bioavailability increases
emtricitabine is not recommended. when it is co-administered with
The most common adverse effects trimethoprim-sulfamethoxazole.
observed in patients receiving Lamivudine and zalcitabine may inhibit the
emtricitabine are headache, insomnia, intracellular phosphorylation of one
nausea, and rash. another; therefore, their concurrent use
In addition, hyperpigmentation of the should be avoided if possible.
palms or soles may be observed (~ 3%),
particularly in African-Americans (up to
13%).

LAMIVUDINE
Lamivudine (3TC) is a cytosine analog
(eFigure 49–3.1) with in vitro activity
against HIV-1 that is synergistic with a
variety of antiretroviral nucleoside
analogs—including zidovudine and
stavudine—against both zidovudine-
sensitive and zidovudine-resistant HIV-1
strains.
As with emtricitabine, lamivudine has STAVUDINE
activity against HBV; therefore,
discontinuation in patients that are co-
The thymidine analog stavudine (d4T) has Like the nucleoside analogs, tenofovir
high oral bioavailability (86%) that is not competitively inhibits HIV reverse
food-dependent. transcriptase and causes chain termination
The serum half-life is 1.1 hours, the after incorporation into DNA.
intracellular half-life is 3.0–3.5 hours, and However, only two rather than three
mean cerebrospinal fluid concentrations intracellular phosphorylations are required
are 55% of those of plasma. for active inhibition of DNA synthesis.
Excretion is by active tubular secretion and Tenofovir is also approved for the
glomerular filtration. treatment of patients with HBV infection.
The major toxicity is a dose-related Tenofovir disoproxil fumarate is a water-
peripheral sensory neuropathy. soluble prodrug of active tenofovir.
The incidence of neuropathy may be The oral bioavailability in fasted patients is
increased when stavudine is administered approximately 25% and increases to 39%
with other potentially neurotoxic drugs after a high-fat meal.
such as didanosine, vincristine, isoniazid, The prolonged serum (12–17 hours) and
or ribavirin, or in patients with advanced intracellular half-lives allow once-daily
immunosuppression. dosing.
Symptoms typically resolve upon Elimination occurs by both glomerular
discontinuation of stavudine; in such cases, filtration and active tubular secretion, and
a reduced dosage may be cautiously dosage adjustment in patients with renal
restarted. insufficiency is recommended.
Other potential adverse effects are Tenofovir is available in several fixed-dose
pancreatitis, arthralgias, and elevation in formulations with emtricitabine, either
serum aminotransferases. alone or in combination with efavirenz,
Lactic acidosis with hepatic steatosis, as rilpivirine, and elvitegravir plus cobicistat.
well as lipodystrophy, appear to occur Based on results of several clinical trials,
more frequently in patients receiving the combination of tenofovir and
stavudine than in those receiving other emtricitabine is now recommended as pre-
NRTI agents. exposure prophylaxis to reduce HIV
Moreover, because the co-administration acquisition in men who have sex with men,
of stavudine and didanosine may increase in heterosexually active men and women,
the incidence of lactic acidosis and and in injection drug users.
pancreatitis, concurrent use should be The primary mutations associated with
avoided. resistance to tenofovir are K65R/N and
This combination has been implicated in K70E.
several deaths in HIV-infected pregnant Gastrointestinal complaints (eg, nausea,
women. diarrhea, vomiting, flatulence) are the most
A rare adverse effect is a rapidly common adverse effects but rarely require
progressive ascending neuromuscular discontinuation of therapy.
weakness. Since tenofovir is formulated with lactose,
Since zidovudine may reduce the these may occur more frequently in
phosphorylation of stavudine, these two patients with lactose intolerance.
drugs should not be used together. Other potential adverse effects include
headache, rash, dizziness, and asthenia.
Cumulative loss of renal function has been
TENOFOVIR observed, possibly increased with
Tenofovir is an acyclic nucleoside concurrent use of boosted PI regimens.
phosphonate (ie, nucleotide) analog of Acute renal failure and Fanconi’s syndrome
adenosine. have also been reported.
For this reason, tenofovir should be used Zidovudine was the first antiretroviral
with caution in patients at risk for renal agent to be approved and has been well
dysfunction. studied.
Serum creatinine levels should be The drug has been shown to decrease the
monitored during therapy and tenofovir rate of clinical disease progression and
discontinued for new proteinuria, prolong survival in HIV-infected
glycosuria, or calculated glomerular individuals.
filtration rate < 30 mL/min. Efficacy has also been demonstrated in the
Tenofovir-associated proximal renal treatment of HIV-associated dementia and
tubulopathy causes excessive renal thrombocytopenia.
phosphate and calcium losses and 1- Studies evaluating the use of zidovudine
hydroxylation defects of vitamin D. during pregnancy, labor, and postpartum
Osteomalacia has been demonstrated in showed significant reductions in the rate
several animal species, and tenofovir use of vertical transmission, and zidovudine
has been an independent risk factor for remains one of the first-line agents for use
bone fracture in some studies. in pregnant women (Table 49–5).
Therefore, monitoring of bone mineral High-level zidovudine resistance is
density should be considered with long- generally seen in strains with three or more
term use in those with risk factors for (or of the five most common mutations: M41L,
known) osteoporosis, as well as in children; D67N, K70R, T215F, and K219Q.
additionally, alternative agents could be However, the emergence of certain
considered in post-menopausal women. mutations that confer decreased
Tenofovir may compete with other drugs susceptibility to one drug (eg, L74V for
that are actively secreted by the kidneys, didanosine and M184V for lamivudine)
such as cidofovir, acyclovir, and may enhance zidovudine susceptibility in
ganciclovir. previously zidovudine-resistant strains.
Concurrent use of atazanavir or Withdrawal of zidovudine may permit the
lopinavir/ritonavir may increase serum reversion of zidovudine-resistant HIV-1
levels of tenofovir (Table 49–4). isolates to the susceptible wild-type
phenotype.
The most common adverse effect of
ZIDOVUDINE zidovudine is myelosuppression, resulting
Zidovudine (azidothymidine; AZT) is a in macrocytic anemia (1–4%) or
deoxythymidine analog that is well neutropenia (2–8%).
absorbed (63%) and distributed to most Gastrointestinal intolerance, headaches,
body tissues and fluids, including the and insomnia may occur but tend to
cerebrospinal fluid, where drug levels are resolve during therapy.
60–65% of those in serum. Lipoatrophy appears to be more common
Although the serum half-life averages 1 in patients receiving zidovudine or other
hour, the intracellular half-life of the thymidine analogs.
phosphorylated compound is 3–4 hours, Less common toxicities include
allowing twice-daily dosing. thrombocytopenia, hyperpigmentation of
Zidovudine is eliminated primarily by renal the nails, and myopathy.
excretion following glucuronidation in the High doses can cause anxiety, confusion,
liver. and tremulousness.
Zidovudine is available in a fixed-dose Increased serum levels of zidovudine may
formulation with lamivudine, either alone occur with concomitant administration of
or in combination with abacavir. probenecid, phenytoin, methadone,
fluconazole, atovaquone, valproic acid,
and lamivudine, either through inhibition latter of which may infrequently be serious
of first-pass metabolism or through (eg, Stevens-Johnson syndrome).
decreased clearance. A further limitation to use of NNRTI agents
Zidovudine may decrease phenytoin as a component of antiretroviral therapy is
levels. their metabolism by the CYP450 system,
Hematologic toxicity may be increased leading to innumerable potential drug-drug
during co-administration of other
interactions (Tables 49–3 and 49–4).
myelosuppressive drugs such as
All NNRTI agents are substrates for CYP3A4
ganciclovir, ribavirin, and cytotoxic agents.
and can act as inducers (nevirapine),
Combination regimens containing
zidovudine and stavudine should be inhibitors (delavirdine), or mixed inducers
avoided due to in vitro antagonism. and inhibitors (efavirenz, etravirine).
Given the large number of non-HIV
medications that are also metabolized by this
NONNUCLEOSIDE REVERSE TRANSCRIPTASE pathway (see Chapter 4), drug-drug
INHIBITORS (NNRTIs) interactions must be expected and looked
for; dosage adjustments are frequently
The NNRTIs bind directly to HIV-1 reverse
required and some combinations are
transcriptase (Figure 49–3), resulting in
contraindicated.
allosteric inhibition of RNA- and DNA-
dependent DNA polymerase activity. DELAVIRDINE
The binding site of NNRTIs is near to but Delavirdine has an oral bioavailability of
distinct from that of NRTIs. about 85%, but this is reduced by antacids
Unlike the NRTI agents, NNRTIs neither or H2 -blockers.
It is extensively bound (~ 98%) to plasma
compete with nucleoside triphosphates nor
proteins and has correspondingly low
require phosphorylation to be active.
cerebrospinal fluid levels.
Baseline genotypic testing is recommended
Serum half-life is approximately 6 hours.
prior to initiating NNRTI treatment because Skin rash occurs in up to 38% of patients
primary resistance rates range from receiving delavirdine; it typically occurs
approximately 2% to 8%. during the first 1–3 weeks of therapy and
NNRTI resistance occurs rapidly with does not preclude rechallenge.
monotherapy and can result from a single However, severe rash such as erythema
mutation. multiforme and Stevens-Johnson
The K103N and Y181C mutations confer syndrome have rarely been reported.
resistance to the first-generation NNRTIs, but Other possible adverse effects are
not to the newer agents (ie, etravirine, headache, fatigue, nausea, diarrhea, and
increased serum aminotransferase levels.
rilpivirine).
Delavirdine has been shown to be
Other mutations (eg, L100I, Y188C, G190A)
teratogenic in rats, causing ventricular
may also confer cross-resistance among the
septal defects and other malformations at
NNRTI class.
dosages not unlike those achieved in
However, there is no cross-resistance humans.
between the NNRTIs and the NRTIs; in fact, Thus, pregnancy should be avoided when
some nucleoside-resistant viruses display taking delavirdine.
hypersusceptibility to NNRTIs. Delavirdine is extensively metabolized by
As a class, NNRTI agents tend to be the CYP3A and CYP2D6 enzymes and also
associated with varying levels of inhibits CYP3A4 and 2C9.
gastrointestinal intolerance and skin rash, the
Therefore, there are numerous potential Other potential adverse reactions are
drug-drug interactions to consider (Tables nausea, vomiting, diarrhea, crystalluria,
49–3 and 49–4). elevated liver enzymes, and an increase in
The concurrent use of delavirdine with total serum cholesterol by 10–20%.
fosamprenavir and rifabutin is not High rates of fetal abnormalities, such as
recommended because of decreased neural tube defects, occurred in pregnant
delavirdine levels. monkeys exposed to efavirenz in doses
Other medications likely to alter roughly equivalent to the human dosage;
delavirdine levels include didanosine, several cases of congenital anomalies have
lopinavir, nelfinavir, and ritonavir. been reported in humans.
Co-administration of delavirdine with Therefore, efavirenz should be avoided in
indinavir or saquinavir prolongs the pregnant women, particularly in the first
elimination half-life of these protease trimester.
inhibitors, thus allowing them to be dosed As both an inducer and an inhibitor of
twice rather than thrice daily. CYP3A4, efavirenz induces its own
metabolism and interacts with the
metabolism of many other drugs (Tables
EFAVIRENZ 49–3 and 49–4).
Efavirenz can be given once daily because Since efavirenz may lower methadone
of its long half-life (40–55 hours). levels, patients receiving these two agents
It is moderately well absorbed following concurrently should be monitored for
oral administration (45%). signs of opioid withdrawal and may
Since toxicity may increase owing to require an increased dose of methadone.
increased bioavailability after a high-fat
meal, efavirenz should be taken on an
empty stomach. ETRAVIRINE
Efavirenz is principally metabolized by Etravirine was designed to be effective
CYP3A4 and CYP2B6 to inactive against strains of HIV that had developed
hydroxylated metabolites; the remainder is resistance to first-generation NNRTIs, due
eliminated in the feces as unchanged drug. to mutations such as K103N and Y181C,
It is highly bound to albumin (~ 99%), and and is recommended for treatment-
cerebrospinal fluid levels range from 0.3% experienced patients that have resistance
to 1.2% of plasma levels. to other NNRTIs.
The principal adverse effects of efavirenz Although etravirine has a higher genetic
involve the central nervous system. barrier to resistance than the other
Dizziness, drowsiness, insomnia, NNRTIs, mutations selected by etravirine
nightmares, and headache tend to usually are associated with resistance to
diminish with continued therapy; dosing at efavirenz, nevirapine, and delavirdine.
bedtime may also be helpful. Etravirine should be taken with a meal to
Psychiatric symptoms such as depression, increase systemic exposure.
mania, and psychosis have been observed It is highly protein-bound and is primarily
and may necessitate discontinuation. metabolized by the liver.
Skin rash has also been reported early in Mean terminal half-life is approximately 41
therapy in up to 28% of patients; the rash hours.
is usually mild to moderate in severity and The most common adverse effects of
typically resolves despite continuation. etravirine are rash, nausea, and diarrhea.
Rarely, rash has been severe or life- The rash is typically mild and usually
threatening. resolves after 1–2 weeks without
discontinuation of therapy.
Rarely, rash has been severe or life- There is no evidence of human
threatening. teratogenicity. However, resistance has
Laboratory abnormalities include been documented after this single dose.
elevations in serum cholesterol, Rash, usually a maculopapular eruption
triglyceride, glucose, and hepatic that spares the palms and soles, occurs in
transaminase levels. up to 20% of patients, usually in the first 4–
Transaminase elevations are more 6 weeks of therapy.
common in patients with HBV or HCV co- Although typically mild and self-limited,
infection. rash is dose-limiting in about 7% of
Etravirine is a substrate as well as an patients.
inducer of CYP3A4 and an inhibitor of Women appear to have an increased
CYP2C9 and CYP2C19; it has many incidence of rash.
therapeutically significant drug-drug When initiating therapy, gradual dose
interactions (Tables 49–3 and 49–4). escalation over 14 days is recommended
Some of the interactions are difficult to to decrease the incidence of rash.
predict. Severe and life-threatening skin rashes
For example, etravirine may decrease have been rarely reported, including
itraconazole and ketoconazole Stevens-Johnson syndrome and toxic
concentrations but increase voriconazole epidermal necrolysis.
concentrations. Nevirapine therapy should be immediately
Etravirine should not be given with other discontinued in patients with severe rash
NNRTIs, unboosted protease inhibitors, and in those with accompanying
atazanavir/ritonavir, constitutional symptoms; since rash may
fosamprenavir/ritonavir, or accompany hepatotoxicity, liver tests
tipranavir/ritonavir. should be assessed.
Symptomatic liver toxicity may occur in up
to 4% of patients, may be severe, and is
NEVIRAPINE more frequent in those with higher
The oral bioavailability of nevirapine is pretherapy CD4 cell counts (ie, > 250
excellent (> 90%) and is not food- cells/mm3 in women and > 400 cells/mm3
dependent. in men), in women, and in those with HBV
The drug is highly lipophilic and achieves or HCV co-infection.
cerebrospinal fluid levels that are 45% of Fulminant, life-threatening hepatitis has
those in plasma. been reported, typically within the first 18
Serum half-life is 25–30 hours. weeks of therapy.
It is extensively metabolized by the CYP3A Other adverse effects include fever,
isoform to hydroxylated metabolites and nausea, headache, and somnolence.
then excreted, primarily in the urine. Nevirapine is a moderate inducer of CYP3A
A single dose of nevirapine (200 mg) is metabolism, resulting in decreased levels
effective in the prevention of transmission of amprenavir, indinavir, lopinavir,
of HIV from mother to newborn when saquinavir, efavirenz, and methadone.
administered to women at the onset of Drugs that induce the CYP3A system, such
labor and followed by a 2 mg/kg oral dose as rifampin, rifabutin, and St. John’s wort,
to the neonate within 3 days after delivery, can decrease levels of nevirapine, whereas
and nevirapine remains one of the those that inhibit CYP3A activity, such as
recommended agents in pregnant women fluconazole, ketoconazole, and
(Table 49–5). clarithromycin, can increase nevirapine
levels.
Since nevirapine may lower methadone depression, headache, insomnia, and
levels, patients receiving these two agents increased serum aminotransferases.
concurrently should be monitored for Increased serum cholesterol, and fat
signs of opioid withdrawal and may redistribution syndrome have been
require an increased dose of methadone. reported. Higher doses have been
associated with QTc prolongation.

RILPIVIRINE
Rilpivirine is recommended only in PROTEASE INHIBITORS (PIs)
treatment-naive patients with HIV-1 RNA
≤100,000 copies/mL, and only in During the later stages of the HIV growth
combination with at least 2 other cycle, thegag and gag-pol gene products are
antiretroviral agents. translated into polyproteins, and these
It is available in a fixed dose formulation become immature budding particles.
with emtricitabine and tenofovir.
The HIV protease is responsible for cleaving
Rilpivirine must be administered with a
these precursor molecules to produce the
meal (preferably high fat or > 400 kcal).
final structural proteins of the mature virion
Its oral bioavailability can be significantly
reduced in the presence of acid lowering core.
agents. By preventing post-translational cleavage of
It should be used with caution with the Gag-Pol polyprotein, protease inhibitors
antacids and H2 -receptor antagonists. (PIs) prevent the processing of viral proteins
Rilpivirine use with proton-pump into functional conformations, resulting in
inhibitors (PPIs) is contraindicated. the production of immature, noninfectious
The drug is highly protein bound and the viral particles (Figure 49–3).
terminal elimination half-life is 50 hours. Unlike the NRTIs, PIs do not need
The E138K substitution emerged most intracellular activation.
frequently during rilpivirine treatment, Specific genotypic alterations that confer
commonly in combination with the M184I
phenotypic resistance are fairly common
substitution. There is cross-resistance with
with these agents, thus contraindicating
other NNRTIs, and the combination of
monotherapy.
rilpivirine with other NNRTIs is not
recommended. Some of the most common mutations
Rilpivirine is primarily metabolized by conferring broad resistance to PIs are
CYP3A4, and drugs that induce or inhibit substitutions at the 10, 46, 54, 82, 84, and 90
CYP3A4 may thus affect the clearance of codons; the number of mutations may
rilpivirine. predict the level of phenotypic resistance.
However, clinically significant drug-drug The I50L substitution emerging during
interactions with other antiretroviral atazanavir therapy has been associated with
agents have not been identified to date. increased susceptibility to other PIs.
Concurrent use of carbamazepine, Darunavir and tipranavir appear to have
dexamethasone, phenobarbital, improved virologic activity in patients
phenytoin, proton pump inhibitors,
harboring HIV-1 resistant to other PIs.
rifabutin, rifampin, rifapentine, and St
As a class, PIs are associated with mild-to-
John’s wort is contraindicated.
moderate nausea, diarrhea, and
Methadone withdrawal may be
precipitated with concurrent usage. dyslipidemia.
The most common adverse effects A syndrome of redistribution and
associated with rilpivirine therapy are rash, accumulation of body fat that results in
central obesity, dorsocervical fat Expert resources about drug-drug
enlargement (buffalo hump), peripheral and interactions should be consulted, as dosage
facial wasting, breast enlargement, and a adjustments are frequently required and
cushingoid appearance has been observed, some combinations are contraindicated.
perhaps less commonly with atazanavir (see It is noteworthy that the potent CYP3A4
below). inhibitory properties of ritonavir are used to
Concurrent increases in triglyceride and low- clinical advantage by having it “boost” the
density lipoprotein levels, along with levels of other PI agents when given in
hyperglycemia and insulin resistance, have combination, thus acting as a
also been noted. pharmacokinetic enhancer rather than an
Abacavir, lopinavir/ritonavir, and antiretroviral agent.
fosamprenavir/ritonavir have been Ritonavir boosting increases drug exposure,
associated with an increased risk of thereby prolonging the drug’s half-life and
cardiovascular disease in some, but not all, allowing reduction in frequency; in addition,
studies. the genetic barrier to resistance is raised.
All PIs may be associated with cardiac
ATAZANAVIR
conduction abnormalities, including PR or QT
Atazanavir (eFigure 49–3.1) is an
interval prolongation or both.
azapeptide PI with a pharmacokinetic
A baseline electrocardiogram and avoidance profile that allows once-daily dosing.
of other agents causing prolonged PR or QT Atazanavir requires an acidic medium for
intervals should be considered. absorption and exhibits pH-dependent
Drug-induced hepatitis and rare severe aqueous solubility; therefore, it should be
hepatotoxicity have been reported to varying taken with meals and separation of
degrees with all PIs; the frequency of hepatic ingestion from acid-reducing agents by at
events is higher with tipranavir/ritonavir than least 12 hours is recommended;
with other PIs. concurrent proton pump inhibitors are
Whether PI agents are associated with bone contraindicated.
loss and osteoporosis after long-term use is Atazanavir is able to penetrate both the
cerebrospinal and seminal fluids.
under investigation.
The plasma half-life is 6–7 hours, which
PIs have been associated with increased
increases to approximately 11 hours when
spontaneous bleeding in patients with
co-administered with ritonavir.
hemophilia A or B; an increased risk of The primary route of elimination is biliary;
intracranial hemorrhage has been reported atazanavir should not be given to patients
in patients receiving tipranavir with ritonavir. with severe hepatic insufficiency.
All of the antiretroviral PIs are extensively Atazanavir is one of the recommended
metabolized by CYP3A4, with ritonavir antiretroviral agents for pregnant women
having the most pronounced inhibitory (Table 49–5).
effect and saquinavir the least. Resistance to atazanavir has been
Some PI agents, such as amprenavir and associated with various known PI
ritonavir, are also inducers of specific CYP mutations as well as with the novel I50L
substitution.
isoforms.
Whereas some atazanavir resistance
As a result, there is enormous potential for
mutations have been associated in vitro
drug-drug interactions with other
with decreased susceptibility to other PIs,
antiretroviral agents and other commonly the I50L mutation has been associated
used medications (Tables 49–3 and 49–4). with increased susceptibility to other PIs.
The most common adverse effects in frequent than with other boosted PI
patients receiving atazanavir are diarrhea regimens) and increases in amylase and
and nausea; vomiting, abdominal pain, hepatic transaminase levels.
headache, peripheral neuropathy, and skin Liver toxicity, including severe hepatitis,
rash may also occur. has been reported in some patients taking
As with indinavir, indirect darunavir; the risk of hepatotoxicity may
hyperbilirubinemia with overt jaundice be higher for persons with HBV, HCV, or
may occur in approximately 10% of other chronic liver disease.
patients, owing to inhibition of the Darunavir contains a sulfonamide moiety
UGT1A1 glucuronidation enzyme. and may cause a hypersensitivity reaction,
Elevation of hepatic enzymes has also particularly in patients with sulfa allergy.
been observed, usually in patients with Darunavir both inhibits and is metabolized
underlying HBV or HCV co-infection. by the CYP3A enzyme system, conferring
Nephrolithiasis has been described in many possible drug-drug interactions
association with atazanavir use, and (Tables 49–3 and 49–4).
prolonged use of boosted atazanavir is In addition, the co-administered ritonavir
associated with cumulative loss of renal is a potent inhibitor of CYP3A and CYP2D6,
function. and an inducer of other hepatic enzyme
In contrast to the other PIs, atazanavir does systems.
not appear to be associated with
dyslipidemia, fat redistribution, or the
metabolic syndrome. FOSAMPRENAVIR
As an inhibitor of CYP3A4 and CYP2C9, the Fosamprenavir is a prodrug of amprenavir
potential for drug-drug interactions with that is rapidly hydrolyzed by enzymes in
atazanavir is great T( ables 49–3 and 49–4). the intestinal epithelium.
Atazanavir AUC is reduced by up to 76% Because of its significantly lower daily pill
when combined with a proton pump burden, fosamprenavir tablets have
inhibitor; thus, this combination is to be replaced amprenavir capsules for adults.
avoided. Fosamprenavir is most often administered
In addition, co-administration of in combination with low-dose ritonavir.
atazanavir with other drugs that inhibit After hydrolysis of fosamprenavir,
UGT1A1, such as irinotecan, may increase amprenavir is rapidly absorbed from the
its levels. gastrointestinal tract, and its prodrug can
Tenofovir and efavirenz should not be co- be taken with or without food.
administered with atazanavir unless However, high-fat meals decrease
ritonavir is added to boost levels. absorption and thus should be avoided.
The plasma half-life is relatively long (7–11
hours).
DARUNAVIR Amprenavir is metabolized in the liver and
Darunavir is licensed as a PI that must be should be used with caution in the setting
co-administered with ritonavir. of hepatic insufficiency.
Darunavir should be taken with meals to The most common adverse effects of
improve bioavailability. fosamprenavir are headache, nausea,
It is highly protein-bound and primarily diarrhea, perioral paresthesias, depression.
metabolized by the liver. Fosamprenavir contains a sulfa moiety and
Symptomatic adverse effects of darunavir may cause a rash in up to 3% of patients,
include diarrhea, nausea, headache, and sometimes severe enough to warrant drug
rash. Laboratory abnormalities include discontinuation.
dyslipidemia (though possibly less
Amprenavir is both an inducer and an and nephrolithiasis due to urinary
inhibitor of CYP3A4 and is contraindicated crystallization of the drug.
with numerous drugs (Tables 49–3 and 49– Nephrolithiasis can occur within days after
4). initiating therapy, with an estimated
The oral solution, which contains incidence of approximately 10%.
propylene glycol, is contraindicated in Consumption of at least 48 ounces of
young children, pregnant women, patients water daily is important to maintain
with renal or hepatic failure, and those adequate hydration.
using metronidazole or disulfiram. Thrombocytopenia, elevations of serum
Also, the oral solutions of amprenavir and aminotransferase levels, nausea, diarrhea,
ritonavir should not be co-administered insomnia, dry throat, dry skin, and indirect
because the propylene glycol in one and hyperbilirubinemia have also been
the ethanol in the other may compete for reported.
the same metabolic pathway, leading to Insulin resistance may be more common
accumulation of either. with indinavir than with the other PIs,
Because the oral solution also contains occurring in 3–5% of patients.
vitamin E at several times the There have also been rare cases of acute
recommended daily dosage, supplemental hemolytic anemia. Since indinavir is an
vitamin E should be avoided. inhibitor of CYP3A4, numerous and
Amprenavir, a sulfonamide, is complex drug interactions can occur
contraindicated in patients with a history (Tables 49–3 and 49–4).
of sulfa allergy. Combination with ritonavir (boosting)
Lopinavir/ritonavir should not be co- allows for twice-daily rather than thrice-
administered with amprenavir owing to daily dosing and eliminates the food
decreased amprenavir and altered restriction associated with use of indinavir.
lopinavir exposures. However, there is potential for an increase
An increased dosage of amprenavir is in nephrolithiasis with this combination
recommended when co-administered with compared with indinavir alone; thus, a high
efavirenz (with or without the addition of fluid intake (1.5–2 L/d) is advised.
ritonavir to boost levels).

LOPINAVIR
INDINAVIR Lopinavir is currently available only in
Indinavir requires an acidic environment combination with ritonavir, which inhibits
for optimum solubility and therefore must the CYP3A-mediated metabolism of
be consumed on an empty stomach or lopinavir, thereby resulting in increased
with a small, low-fat, low-protein meal for exposure to lopinavir.
maximal absorption (60–65%). In addition to improved patient
The serum half-life is 1.5–2 hours, protein compliance due to reduced pill burden,
binding is approximately 60%, and the lopinavir/ritonavir is generally well
drug has a high level of cerebrospinal fluid tolerated.
penetration (up to 76% of serum levels). Lopinavir is highly protein bound (98–
Excretion is primarily fecal. 99%), and its half-life is 5–6 hours. It is
An increase in AUC by 60% and in half-life extensively metabolized by CYP3A, which
to 2.8 hours in the setting of hepatic is inhibited by ritonavir.
insufficiency necessitates dose reduction. Serum levels of lopinavir may be increased
The most common adverse effects of in patients with hepatic impairment.
indinavir are indirect hyperbilirubinemia
Lopinavir/ritonavir is one of the Nelfinavir is an inhibitor of the CYP3A
recommended antiretroviral agents for use system, and multiple drug interactions
in pregnant women (Table 49–5). may occur (Tables 49–3 and 49–4).
The most common adverse effects of An increased dosage of nelfinavir is
lopinavir are diarrhea, abdominal pain, recommended when co-administered with
nausea, vomiting, and asthenia. rifabutin (with a decreased dose of
Ritonavir-boosted lopinavir may be more rifabutin), whereas a decrease in saquinavir
commonly associated with gastrointestinal dose is suggested with concurrent
adverse events than other PIs. nelfinavir.
Elevations in serum cholesterol and Co-administration with efavirenz should
triglycerides are common. be avoided due to decreased nelfinavir
Prolonged use of boosted lopinavir is levels.
associated with cumulative loss of renal
function, and lopinavir use has been an
independent risk factor for bone fracture RITONAVIR
in some (but not all) studies. Ritonavir (eFigure 49–3.1) has a high
Potential drug-drug interactions are bioavailability (about 75%) that increases
extensive (Tables 49–3 and 49–4). with food. It is 98% protein-bound and has
Increased dosage of lopinavir/ritonavir is a serum half-life of 3–5 hours.
recommended when co-administered with Metabolism to an active metabolite occurs
efavirenz or nevirapine, which induce via the CYP3A and CYP2D6 isoforms;
lopinavir metabolism. excretion is primarily in the feces.
Concurrent use of fosamprenavir should Caution is advised when administering the
be avoided owing to altered exposure to drug to persons with impaired hepatic
lopinavir with decreased levels of function.
amprenavir. Ritonavir is one of the recommended
Also, concomitant use of antiretroviral agents for use in pregnant
lopinavir/ritonavir and rifampin is women (Table 49–5).
contraindicated due to an increased risk Potential adverse effects of ritonavir,
for hepatotoxicity. particularly when administered at full
Since the oral solution of dosage, are gastrointestinal disturbances,
lopinavir/ritonavir contains alcohol, paresthesias (circumoral or peripheral),
concurrent disulfiram and metronidazole elevated serum aminotransferase levels,
are contraindicated. altered taste, headache, and elevations in
serum creatine kinase.
Nausea, vomiting, diarrhea, or abdominal
NELFINAVIR pain typically occur during the first few
Nelfinavir has high absorption in the fed weeks of therapy but may diminish over
state (70–80%), undergoes metabolism by time or if the drug is taken with meals.
CYP3A, and is excreted primarily in the Dose escalation over 1–2 weeks is
feces. recommended to decrease the dose-
The plasma half-life in humans is 3.5–5 limiting side effects.
hours, and the drug is more than 98% Ritonavir is a potent inhibitor of CYP3A4,
protein-bound. resulting in many potential drug
The most common adverse effects interactions (Tables 49–3 and 49–4).
associated with nelfinavir are diarrhea and However, this characteristic has been used
flatulence. to great advantage when ritonavir is
Diarrhea often responds to antidiarrheal administered in low doses (100–200 mg
medications but can be dose-limiting. twice daily) in combination with any of the
other PI agents, in that increased blood Saquinavir is subject to extensive first-pass
levels of the latter agents permit lower or metabolism by CYP3A4 and functions as a
less frequent dosing (or both) with greater CYP3A4 inhibitor as well as a substrate;
tolerability as well as the potential for thus, there are many potential drug-drug
greater efficacy against resistant virus. interactions (Tables 49–3 and 49–4).
Therapeutic levels of digoxin and A decreased dose of saquinavir is
theophylline should be monitored when recommended when co-administered with
co-administered with ritonavir owing to a nelfinavir. Increased saquinavir levels when
likely increase in their concentrations. co-administered with omeprazole
The concurrent use of saquinavir and necessitate close monitoring for toxicities.
ritonavir is contraindicated due to an Digoxin levels may increase if co-
increased risk of QT prolongation (with administered with saquinavir and should
torsades de pointes arrhythmia) and PR therefore be monitored.
interval prolongation. Liver tests should be monitored if
saquinavir is co-administered with
delavirdine or rifampin.
SAQUINAVIR
In its original formulation as a hard gel
capsule, oral saquinavir was poorly TIPRANAVIR
bioavailable (only about 4% after food). Tipranavir is a newer PI indicated for use in
However, reformulation of saquinavir for treatment-experienced patients who
once-daily dosing in combination with harbor strains resistant to other PI agents.
low-dose ritonavir has both improved It is used in combination with ritonavir to
antiviral efficacy and decreased achieve effective serum levels.
gastrointestinal adverse effects. Bioavailability is poor but is increased
A previous formulation of saquinavir in when taken with a high-fat meal.
soft gel capsules is no longer available. The drug is metabolized by the liver
Saquinavir should be taken within 2 hours microsomal system and is contraindicated
after a fatty meal for enhanced absorption. in patients with hepatic insufficiency.
Saquinavir is 97% protein-bound, and Tipranavir contains a sulfonamide moiety
serum half-life is approximately 2 hours. and should not be administered to
Saquinavir has a large volume of patients with known sulfa allergy.
distribution, but penetration into the The most common adverse effects of
cerebrospinal fluid is negligible. tipranavir are diarrhea, nausea, vomiting,
Excretion is primarily in the feces. and abdominal pain.
Reported adverse effects include An urticarial or maculopapular rash is more
gastrointestinal discomfort (nausea, common in women and may be
diarrhea, abdominal discomfort, accompanied by systemic symptoms or
dyspepsia) and rhinitis. desquamation.
When administered in combination with Liver toxicity, including life-threatening
low-dose ritonavir, there appears to be hepatic decompensation, has been
less dyslipidemia or gastrointestinal observed and may be more common than
toxicity than with some of the other with other PIs, particularly in patients with
boosted PI regimens. chronic HBV or HCV infection.
However, the concurrent use of saquinavir Tipranavir should be discontinued in
and ritonavir may confer an increased risk patients who have increased serum
of QT prolongation (with torsades de transaminase levels that are more than 10
pointes arrhythmia) and PR interval times the upper limit of normal or more
prolongation.
than 5 times normal in combination with ENFUVIRTIDE
increased serum bilirubin. Enfuvirtide is a synthetic 36-amino-acid
Because of an increased risk for peptide fusion inhibitor that blocks HIV
intracranial hemorrhage in patients entry into the cell (Figure 49–3).
receiving tipranavir/ritonavir, the drug Enfuvirtide binds to the gp41 subunit of
should be avoided in patients with head the viral envelope glycoprotein,
trauma or bleeding diathesis. preventing the conformational changes
Other potential adverse effects include required for the fusion of the viral and
depression, elevation in amylase, and cellular membranes.
decreased white blood cell count. It is administered in combination with
Tipranavir both inhibits and induces the other antiretroviral agents in treatment-
CYP3A4 system. When used in experienced patients with evidence of viral
combination with ritonavir, its net effect is replication despite ongoing antiretroviral
inhibition. Tipranavir also induces P- therapy.
glycoprotein transporter and thus may Enfuvirtide, which must be administered
alter the disposition of many other drugs by subcutaneous injection, is the only
(Table 49–4). Concurrent administration of parenterally administered antiretroviral
tipranavir with fosamprenavir or saquinavir agent.
should be avoided owing to decreased Metabolism appears to be by proteolytic
blood levels of the latter drugs. hydrolysis without involvement of the
Tipranavir/ritonavir may also decrease CYP450 system.
serum levels of valproic acid and Elimination half-life is 3.8 hours.
omeprazole. Levels of lovastatin, Resistance to enfuvirtide can result from
simvastatin, atorvastatin, and rosuvastatin mutations in gp41; the frequency and
may be increased, increasing the risk for significance of this phenomenon are being
rhabdomyolysis and myopathy. investigated.
However, enfuvirtide lacks cross-resistance
with the other currently approved
ENTRY INHIBITORS antiretroviral drug classes.
The most common adverse effects
The process of HIV-1 entry into host cells is associated with enfuvirtide therapy are
complex; each step presents a potential local injection site reactions, consisting of
target for inhibition. painful erythematous nodules.
Viral attachment to the host cell entails Although frequent, these are typically
binding of the viral envelope glycoprotein mild-to-moderate and rarely lead to
complex gp160 (consisting of gp120 and discontinuation.
gp41) to its cellular receptor CD4. Other side effects may include insomnia,
This binding induces conformational headache, dizziness, and nausea.
Hypersensitivity reactions may rarely
changes in gp120 that enable access to the
occur, are of varying severity, and may
chemokine receptors CCR5 or CXCR4.
recur on rechallenge.
Chemokine receptor binding induces further
Eosinophilia is the primary laboratory
conformational changes in gp120, allowing abnormality seen with enfuvirtide
exposure to gp41 and leading to fusion of administration.
the viral envelope with the host cell No drug-drug interactions have been
membrane and subsequent entry of the viral identified that would require the alteration
core into the cellular cytoplasm. of the dosage of concomitant antiretroviral
or other drugs.
MARAVIROC the fusion inhibitor enfuvirtide. However,
Maraviroc (eFigure 49–3.1) is approved for emergence of CXCR4 virus (either
use in combination with other previously undetected or newly
antiretroviral agents in treatment- developed) appears to be a more common
experienced adult patients infected with cause of virologic failure than the
only CCR5-tropic HIV-1 detectable who development of resistance mutations.
are resistant to other antiretroviral agents. Maraviroc is a substrate for CYP3A4 and
Maraviroc binds specifically and selectively therefore requires adjustment in the
to the host protein CCR5, one of two presence of drugs that interact with these
chemokine receptors necessary for enzymes (Tables 49–3 and 49–4).
entrance of HIV into CD4+ cells. It is also a substrate for P-glycoprotein,
Since maraviroc is active against HIV that which limits intracellular concentrations of
uses the CCR5 co-receptor exclusively, and the drug.
not against HIV strains with CXCR4, dual, The dosage of maraviroc must be
or mixed tropism, co-receptor tropism decreased if it is co-administered with
should be determined by specific testing strong CYP3A inhibitors (eg, delavirdine,
before maraviroc is started, using the ketoconazole, itraconazole, clarithromycin,
enhanced sensitivity tropism assay. or any protease inhibitor other than
Substantial proportions of patients, tipranavir) and must be increased if
particularly those with advanced HIV coadministered with CYP3A inducers (eg,
infection, are likely to have virus that is not efavirenz, etravirine, rifampin,
exclusively CCR5-tropic. carbamazepine, phenytoin, or St. John’s
The absorption of maraviroc is rapid but wort).
variable, with the time to maximum Potential adverse effects of maraviroc
absorption generally being 1–4 hours after include cough, upper respiratory tract
ingestion of the drug. infections, muscle and joint pain, diarrhea,
Most of the drug (≥ 75%) is excreted in the sleep disturbance, and elevations in serum
feces, whereas approximately 20% is aminotransferases.
excreted in urine. Hepatotoxicity has been reported, which
The recommended dose of maraviroc may be preceded by a systemic allergic
varies according to renal function and the reaction (ie, pruritic rash, eosinophilia, or
concomitant use of CYP3A inducers or elevated IgE); discontinuation of maraviroc
inhibitors. should be prompt if this constellation
Maraviroc is contraindicated in patients occurs.
with severe or end-stage renal impairment Also, caution should be used in patients
who are taking concurrent CYP3A with pre-existing liver dysfunction or who
inhibitors or inducers, and caution is are co-infected with HBV or HCV.
advised when used in patients with Myocardial ischemia and infarction have
preexisting hepatic impairment and in been observed in patients receiving
those co-infected with HBV or HCV. maraviroc; therefore caution is advised in
Maraviroc has excellent penetration into patients at increased cardiovascular risk.
the cervicovaginalfluid, with levels almost There has been concern that blockade of
four times higher than the corresponding the chemokine CCR5 receptor—a human
concentrations in blood plasma. protein—may result in decreased immune
Resistance to maraviroc is associated with surveillance, with a subsequent increased
one or more mutations in the V3 loop of risk of malignancy (eg, lymphoma) or
gp120. infection.
There appears to be no cross-resistance To date, however, there has been no
with drugs from any other class, including evidence of an increased risk of either
malignancy or infection in patients Dolutegravir is primarily metabolized via
receiving maraviroc. UGT1A1 with some contribution from
CYP3A.
Therefore, drug-drug interactions may
INTEGRASE STRAND TRANSFER INHIBITORS occur (Table 49–4).
(INSTIs) Co-administration with the metabolic
inducers phenytoin, phenobarbital,
This class of agents binds integrase, a viral carbamazepine, and St. John’s wort should
enzyme essential to the replication of both be avoided.
HIV-1 and HIV-2. Dolutegravir inhibits the renal organic
By doing so, it inhibits strand transfer, the cation transporter OCT2, thereby
third and final step of provirus integration, increasing plasma concentrations of drugs
thus interfering with the integration of eliminated via OCT2 such as dofetilide and
reverse-transcribed HIV DNA into the metformin.
For this reason, co-administration with
chromosomes of host cells (Figure 49–3).
dofetilide is contraindicated and close
As a class, these agents tend to be well
monitoring, with potential for dose
tolerated, with headache and gastrointestinal
adjustment, is recommended for co-
effects being the most commonly reported administration with metformin.
adverse events. Current evidence suggests that
Other nervous system (including dolutegravir retains activity against some
neuropsychiatric) effects are often reported viruses resistant to both raltegravir and
but are milder and less frequent than with elvitegravir.
efavirenz. The most common adverse reactions
Limited data suggest that effects upon lipid associated with dolutegravir are insomnia
metabolism are favorable compared with and headache.
efavirenz and PIs, with more variable findings Hypersensitivity reactions characterized by
for elvitegravir than raltegravir and rash, constitutional findings, and
sometimes organ dysfunction, including
dolutegravir due to co-administration with
liver injury, have been reported and may
the boosting agent cobicistat.
be life-threatening.
Rare and severe events include systemic
The drug should be discontinued
hypersensitivity reactions and immediately if this occurs and not
rhabdomyolysis. restarted.
Other reported side effects include
DOLUTEGRAVIR
elevations in serum aminotransferases and
Dolutegravir may be taken with or without
the fat redistribution syndrome.
food.
The absolute oral bioavailability has not
been established. ELVITEGRAVIR
Dolutegravir should be taken 2 hours Elvitegravir requires boosting with an
before or 6 hours after taking cation- additional drug, such as cobicistat (a
containing antacids or laxatives, sucralfate, pharmacokinetic enhancer that inhibits
oral iron supplements, oral calcium CYP3A4 as well as certain intestinal
supplements, or buffered medications. transport proteins) or ritonavir.
The terminal half-life is approximately 14 Elvitegravir is therefore available only as a
hours. component of a fixed-dose combination,
with cobicistat, emtricitabine, and
tenofovir.
The combined formulation should be single point mutation (eg, at codons 148
taken with food. or 155).
Cobicistat can inhibit renal tubular The low genetic barrier to resistance
secretion of creatinine, causing increases emphasizes the importance of
in serum creatinine that may not be combination therapies and of adherence.
clinically significant; in the fixed-dose Integrase mutations are not expected to
formulation it may be difficult to affect sensitivity to other classes of
distinguish between cobicistat effect and antiretroviral agents.
tenofovir-induced nephrotoxicity. Potential adverse effects of raltegravir
The recommendation is that the fixed- include insomnia, headache, dizziness,
dose combination diarrhea, nausea, fatigue, and muscle
elvitegravir/cobicistat/tenofovir/emtricita aches. Increases in pancreatic amylase,
bine should not be initiated in patients serum aminotransferases, and creatine
with calculated creatinine clearance < 70 kinase (with rhabdomyolysis) may occur.
mL/min and should be discontinued in Severe, potentially life-threatening and
those with creatinine clearance < 50 fatal skin reactions have been reported,
mL/min; discontinuation should be including Stevens-Johnson syndrome,
considered if the serum creatinine hypersensitivity reaction, and toxic
increases by 0.4 mg/dL or more. epidermal necrolysis.

RALTEGRAVIR ANTIHEPATITIS AGENTS


Absolute bioavailability of the
pyrimidinone analog raltegravir has not INTERFERON ALFA
been established but does not appear to
be food-dependent. Interferons are host cytokines that exert
The drug does not interact with the complex antiviral, immunomodulatory, and
cytochrome P450 system butis antiproliferative actions (see Chapter 55) and
metabolized by glucuronidation, some have proven useful in both HBV and
particularly UGT1A1. HCV.
Inducers or inhibitors of UGT1A1 may Interferon alfa appears to function by
affect serum levels of raltegravir. induction of intracellular signals following
For example, since concurrent use of binding to specific cell membrane receptors,
rifampin substantially decreases raltegravir resulting in inhibition of viral penetration,
concentrations, the dose of raltegravir
translation, transcription, protein processing,
should be increased.
maturation, and release, as well as increased
Since polyvalent cations (eg, magnesium,
host expression of major histocompatibility
calcium, and iron) may bind integrase
inhibitors and interfere with their activity, complex antigens, enhanced phagocytic
antacids should be used cautiously and activity of macrophages, and augmentation
ingestion separated by at least 4 hours of the proliferation and survival of cytotoxic
from raltegravir. T T cells.
he chewable tablets may contain Injectable preparations of interferon alfa are
phenylalanine, which can be harmful to available for treatment of both HBV and HCV
patients with phenylketonuria. infections (Table 49–6).
Although virologic failure has been Interferon alfa-2a and interferon alfa-2b may
uncommon in clinical trials of raltegravir to be administered either subcutaneously or
date, in vitro resistance requires only a intramuscularly; half-life is 2–5 hours,
depending on the route of administration.
Alfa interferons are filtered at the glomerulus ischemic cardiac disease, severe renal
and undergo rapid proteolytic degradation insufficiency, and cytopenia.
during tubular reabsorption, such that Alfa interferons are abortifacient in primates
detection in the systemic circulation is and should not be administered in
negligible. pregnancy.
Liver metabolism and subsequent biliary Potential drug-drug interactions include
excretion are considered minor pathways. increased theophylline and methadone
The use of pegylated (polyethylene glycol- levels.
complexed) interferon alfa-2a and pegylated Co-administration with didanosine is not
interferon alfa-2b results in slower clearance, recommended because of a risk of hepatic
longer terminal half-lives, and steadier failure, and co-administration with
concentrations, thus allowing for less zidovudine may exacerbate cytopenias.
frequent dosing. The polyethylene glycol molecule is a
Renal elimination accounts for about 30% of nontoxic polymer that is readily excreted in
clearance, and clearance is approximately the urine.
halved in subjects with impaired renal
TREATMENT OF HEPATITIS B VIRUS INFECTION
function; dosage must therefore be adjusted.
The adverse effects of interferon alfa include The goals of chronic HBV therapy are the
a flu-like syndrome (ie, headache, fevers, suppression of HBV DNA to undetectable
chills, myalgias, and malaise) that typically levels, seroconversion of HBeAg (or more
occurs within 6 hours after dosing; this rarely, HBsAg) from positive to negative, and
syndrome occurs in more than 30% of reduction in elevated hepatic transaminase
patients during the first week of therapy and levels.
tends to resolve upon continued These end points are correlated with
administration. improvement in necroinflammatory disease,
Transient hepatic enzyme elevations may a decreased risk of hepatocellular carcinoma
occur in the first 8–12 weeks of therapy and and cirrhosis, and a decreased need for liver
appear to be more common in responders. transplantation.
Potential adverse effects during chronic All of the currently licensed therapies achieve
therapy include neurotoxicities (mood these goals.
disorders, depression, somnolence, However, because current therapies suppress
confusion, seizures), myelosuppression, HBV replication without eradicating the virus,
profound fatigue, weight loss, rash, cough, initial responses may not be durable.
myalgia, alopecia, tinnitus, reversible hearing The covalently closed circular (ccc) viral DNA
loss, retinopathy, pneumonitis, and possibly exists in stable form indefinitely within the
cardiotoxicity. cell, serving as a reservoir for HBV
Induction of autoantibodies may occur, throughout the life of the cell and resulting
causing exacerbation or unmasking of in the capacity to reactivate.
autoimmune disease (particularly thyroiditis). Relapse is more common in patients co-
Contraindications to interferon alfa therapy infected with HBV and hepatitis D virus.
include hepatic decompensation, As of 2013 seven drugs were approved for
autoimmune disease, and history of cardiac treatment of chronic HBV infection in the
arrhythmia. United States: five oral
Caution is advised in the setting of nucleoside/nucleotide analogs
psychiatric disease, epilepsy, thyroid disease, (lamivudine, adefovir dipivoxil, tenofovir,
entecavir, telbivudine) and two injectable ADEFOVIR DIPIVOXIL
interferon drugs (interferon alfa-2b,
Although initially and abortively developed
pegylated interferon alfa-2a) (Table 49–
for treatment of HIV infection, adefovir
6).
dipivoxil gained approval, at lower and less
toxic doses, for treatment of HBV infection.
Adefovir dipivoxil is the diester prodrug of
adefovir, an acyclic phosphonated adenine
nucleotide analog (eFigure 49–4.1).
It is phosphorylated by cellular kinases to
the active diphosphate metabolite and then
competitively inhibits HBV DNA polymerase
and causes chain termination after
The use of interferon has been supplanted by
incorporation into viral DNA.
long-acting pegylated interferon, allowing
Adefovir is active in vitro against a wide
once-weekly rather than daily or thrice-
range of DNA and RNA viruses, including
weekly dosing.
HBV, HIV, and herpesviruses.
In general, nucleoside/nucleotide analog
Oral bioavailability of adefovir dipivoxil is
therapies have better tolerability and
about 59% and is unaffected by meals; it is
produce a higher response rate than the
rapidly and completely hydrolyzed to the
interferons and are now considered the first
parent compound by intestinal and blood
line of therapy.
esterases.
Combination therapies may reduce the
Protein binding is low (< 5%).
development of resistance.
The intracellular half-life of the diphosphate
The optimal duration of therapy remains
is prolonged, ranging from 5 to 18 hours in
unknown. Several anti-HBV agents have anti-
various cells; this makes once-daily dosing
HIV activity as well, including tenofovir,
feasible.
lamivudine, and adefovir dipivoxil.
Adefovir is excreted by a combination of
Emtricitabine, an NRTI used in HIV infection,
glomerular filtration and active tubular
has resulted in excellent biochemical,
secretion and requires dose adjustment for
virologic, and histologic improvement in
renal dysfunction; however, it may be
patients with chronic HBV infection, although
administered to patients with
it is not approved for this indication.
decompensated liver disease.
Agents with dual HBV and HIV activity are
Of the oral agents, adefovir may be slower to
particularly useful as part of a first-line
suppress HBV DNA levels and the least likely
regimen in co-infected patients.
to induce HBeAg seroconversion.
However, it is important to note that acute
Emergence of resistance is 20% to 30% after
exacerbation of hepatitis may occur upon
5 years of use.
discontinuation or interruption of these
Naturally occurring (ie, primary) adefovir-
agents; this may be severe or even fatal.
resistant rt233 HBV mutants have been
described.
There is no cross-resistance between
adefovir and lamivudine or entecavir.
Adefovir is well tolerated.
A dose-dependent nephrotoxicity,
manifested by increased serum creatinine
and decreased serum phosphorous, may more likely in the setting of lamivudine
occur and is more common with use of resistance.
higher doses (30-60 mg/d) or pre-existing Although selection of resistant isolates
azotemia. with the S202G mutation has been
Other potential adverse effects are headache, documented during therapy, clinical
diarrhea, asthenia, and abdominal pain. resistance is rare (< 1% at 4 years).
Entecavir has weak anti-HIV activity and
As with other NRTI agents, lactic acidosis and
can induce development of the M184V
hepatic steatosis are considered a risk owing
variant in HBV/HIV co-infected patients,
to mitochondrial dysfunction.
resulting in resistance to emtricitabine and
Pivalic acid, a by-product of adefovir lamivudine.
metabolism, can esterify free carnitine and Entecavir is well tolerated.
result in decreased carnitine levels. Potential adverse events are headache,
However, it is not necessary to administer fatigue, dizziness, nausea, rash, and fever.
carnitine supplementation with the low Lung adenomas and carcinomas in mice,
doses used to treat patients with HBV (10 hepatic adenomas and carcinomas in rats
mg/d). and mice, vascular tumors in mice, and
Severe acute exacerbations of hepatitis have brain gliomas and skin fibromas in rats
been reported in up to 25% of patients who have been observed at varying exposures.
Co-administration of entecavir with drugs
discontinued adefovir.
that reduce renal function or compete for
Adefovir is embryotoxic in rats at high doses
active tubular secretion may increase
and is genotoxic in preclinical studies.
serum concentrations of either entecavir
ENTECAVIR or the co-administered drug.
Entecavir is an orally administered
guanosine nucleoside analog. that
LAMIVUDINE
competitively inhibits all three functions of
The pharmacokinetics of lamivudine are
HBV DNA polymerase, including base
described earlier in this chapter (see
priming, reverse transcription of the
section, Nucleoside and Nucleotide
negative strand, and synthesis of the
Reverse Transcriptase Inhibitors).
positive strand of HBV DNA.
The more prolonged intracellular half-life
Oral bioavailability approaches 100% but
in HBV cell lines (17–19 hours) than in HIV-
is decreased by food; therefore, entecavir
infected cell lines (10.5–15.5 hours) allows
should be taken on an empty stomach.
for lower doses and less frequent
The intracellular half-life of the active
administration.
phosphorylated compound is 15 hours
Lamivudine can be safely administered to
and plasma half-life is prolonged at 128-
patients with decompensated liver disease.
149 hours, allowing once-daily dosing.
Prolonged treatment has been shown to
It is excreted by the kidney, undergoing
decrease clinical progression of HBV, as
both glomerular filtration and net tubular
well as development of hepatocellular
secretion.
cancer by approximately 50%.
Suppression of HBV DNA levels was
Also, lamivudine has been effective in
greater with entecavir than with
preventing vertical transmission of HBV
lamivudine or adefovir in comparative
from mother to newborn when given in the
trials Entecavir appears to have a higher
last 4 weeks of gestation.
barrier to the emergence of resistance
Lamivudine inhibits HBV DNA polymerase
than lamivudine but resistance may be
and HIV reverse transcriptase by
competing with deoxycytidine
triphosphate for incorporation into the occur in up to 22% of patients with
viral DNA, resulting in chain termination. durations of therapy exceeding 1 year, and
Although lamivudine initially results in may result in virologic rebound.
rapid and potent virus suppression, Telbivudine is not effective in patients with
chronic therapy is limited by the lamivudine-resistant HBV.
emergence of lamivudine-resistant HBV Adverse effects are mild; they include
isolates (eg, L180M or M204I/V), estimated fatigue, headache, cough, nausea,
to occur in 15–30% of patients at 1 year diarrhea, rash, and fever.
and 70% at 5 years of therapy. Both uncomplicated myalgia and
Resistance has been associated with flares myopathy have been reported, concurrent
of hepatitis and progressive liver disease. with increased creatine kinase levels, as
Cross-resistance between lamivudine and has peripheral neuropathy.
emtricitabine or entecavir may occur; As with other nucleoside analogs, lactic
however, adefovir and tenofovir maintain acidosis and severe hepatomegaly with
activity against lamivudine-resistant steatosis may occur during therapy as well
strains of HBV. as flares of hepatitis after discontinuation.
In the doses used for HBV infection,
lamivudine has an excellent safety profile.
Headache, nausea, diarrhea, dizziness, TENOFOVIR
myalgia, and malaise are rare. Tenofovir, a nucleotide analog of
Co-infection with HIV may increase the risk adenosine in use as an antiretroviral agent,
of pancreatitis. has potent activity against HBV.
The characteristics of tenofovir are
described earlier in this chapter.
TELBIVUDINE Tenofovir maintains activity against
Telbivudine is a thymidine nucleoside lamivudine- and entecavir-resistant
analog with activity against HBV DNA hepatitis virus isolates but has reduced
polymerase. activity against adefovir-resistant strains.
It is phosphorylated by cellular kinases to Although similar in structure to adefovir
the active triphosphate form, which has an dipivoxil, comparative trials show a higher
intracellular half-life of 14 hours. rate of virologic response and histologic
The phosphorylated compound improvement, and a lower rate of
competitively inhibits HBV DNA emergence of resistance to tenofovir in
polymerase, resulting in incorporation into patients with chronic HBV infection.
viral DNA and chain termination. The most common adverse effects of
It is not active in vitro against HIV-1. Oral tenofovir in patients with HBV infection are
bioavailability is unaffected by food. nausea, abdominal pain, diarrhea,
Plasma protein-binding is low (3%) and dizziness, fatigue, and rash; other potential
distribution wide. adverse effects are those listed earlier.
The serum half-life is approximately 15
hours and excretion is renal.
There are no known metabolites and no TREATMENT OF HEPATITIS C INFECTION
known interactions with the CYP450
system or other drugs. In contrast to the treatment of patients with
Telbivudine induced greater rates of chronic HBV infection, the primary goal of
virologic response than either lamivudine treatment in patients with HCV infection is
or adefovir in comparative trials. viral eradication.
However, emergence of resistance, In clinical trials, the primary efficacy end
typically due to the M204I mutation, may point is typically achievement of sustained
viral response (SVR), defined as the absence 45 to 70% among patients with any of the
of detectable viremia 24 weeks after other genotypes.
completion of therapy. A genetic variant near the gene encoding
SVR is associated with improvement in liver interferon-lambda-3 (IL28B rs12979860) is a
histology, reduction in risk of hepatocellular strong predictor of response to
carcinoma, and, occasionally, with regression peginterferon alfa and ribavirin.
of cirrhosis as well. However, the recent advent of NS3/4A
Late relapse occurs in less than 5% of protease inhibitors and the NS5B polymerase
patients who achieve SVR. inhibitors is changing the face of chronic
In acute hepatitis C, the rate of clearance of HCV therapy.
the virus without therapy is estimated at 15– Administration of boceprevir, simeprevir, or
30%. In one (uncontrolled) study, treatment telaprevir, in combination with peginterferon
of acute infection with interferon alfa-2b, in and ribavirin, dramatically increased the rate
doses higher than those used for chronic of viral clearance in patients with HCV
hepatitis C, resulted in a sustained rate of genotype 1; sofosbuvir is effective against
clearance of 95% at 6 months. HCV genotypes 1, 2, 3, and 4.
Therefore, if HCV RNA testing documents Although all four of these new agents are
persistent viremia 12 weeks after initial licensed to be administered in combination
seroconversion, antiviral therapy is with peginterferon and ribavirin, recent
recommended. results of clinical trials have provided
Treatment of patients with chronic HCV evidence that one or more of them may be
infection is recommended for those with an effective in interferon- and ribavirin-free
increased risk for progression to cirrhosis. regimens.
The parameters for selection are complex. In
POLYMERASE INHIBITORS
those who are to be treated, the traditional
standard treatment is once-weekly pegylated
Sofosbuvir
interferon alfa in combination with daily oral
Sofosbuvir is a nucleotide analog that
ribavirin. inhibits the HCV NS5B RNA-dependent
Pegylated interferon alfa-2a and -2b have RNA polymerase in patients infected with
replaced their unmodified interferon alfa HCV genotype 1, 2, 3, or 4.
counterparts because of superior efficacy in It is administered once daily, with or
combination with ribavirin, regardless of without food, in combination with
genotype. peginterferon alfa and ribavirin, for a total
It is also clear that combination therapy with of 12–24 weeks (the longer duration is
oral ribavirin is more effective than recommended in patients infected with
monotherapy with either interferon or HCV genotype 3).
ribavirin alone. Very high cure rates are reported but the
drug is extraordinarily expensive.
Therefore, monotherapy with pegylated
Sofosbuvir is 61–65% bound to plasma
interferon alfa is recommended only in
proteins and is metabolized in the liver to
patients who cannot tolerate ribavirin.
form the active nucleoside analog
Interferon plus ribavirin therapy is active triphosphate GS-461203.
against all genotypes of HCV infection, with Elimination is by renal clearance, and
SVR rates of 70 to 80% among patients with safety has not been established in patients
HCV genotype 2 or 3 infection and rates of with severe renal insufficiency.
Sofosbuvir is a substrate of drug Since boceprevir, simeprevir, and telaprevir
transporter P-gp; therefore, potent P-gp are always co-administered with ribavirin,
inducers in the intestine should not be co- their use in pregnant women and in men with
administered. pregnant partners is contraindicated.
Commonly reported adverse effects are
fatigue and headache. Boceprevir
Boceprevir therapy is initiated after the
administration of peginterferon and
PROTEASE INHIBITORS ribavirin therapy for 4 weeks. The duration
of therapy is dependent on the
Three oral protease NS3/4A inhibitors have achievement of undetectable virus.
recently become available for the treatment Boceprevir should be taken with food to
of HCV genotype 1 infection, in combination maximize absorption. It is ~75% protein-
with peginterferon and ribavirin: boceprevir, bound and has a mean plasma halflife of
simeprevir, and telaprevir. approximately 3.4 hours. Boceprevir is
These agents inhibit HCV replication directly metabolized by the aldo-keto-reductase
and CYP3A4/5 pathways and is an inhibitor
by binding to the NS3/4A protease that
of CYP3A4/5 and P-glycoprotein
cleaves HCV-encoded polyproteins (Figure
transporter. Co-administration of
49–4).
boceprevir with numerous drugs is
Of concern is the enhanced toxicity when
contraindicated, including carbamazepine,
used in combination with peginterferon and phenobarbital, phenytoin, rifampin, ergot
ribavirin, the high potential for drug-drug derivatives, cisapride, lovastatin,
interactions, and the low genetic barrier to simvastatin, St. John’s wort, drospirenone,
resistance, which may develop as early as 4 alfuzosin, sildenafil or tadalafil when used
days after initiation of therapy when for pulmonary hypertension, pimozide,
administered as monotherapy. triazolam, midazolam, and efavirenz.
Use of these agents in the treatment of other The most commonly reported adverse
HCV genotypes is not recommended. effects associated with boceprevir therapy
Cross-resistance is expected among NS3/4A are fatigue, anemia, neutropenia, nausea,
headache, and dysgeusia.
protease inhibitors.
Rates of anemia are higher in patients
All three agents are inhibitors and substrates
taking boceprevir with peginterferon and
of CYP3A inhibitors.
ribavirin than in those taking
Drug-drug interactions are to be expected peginterferon and ribavirin alone (~ 50%
with many concurrent agents, particularly the vs 25%, respectively); rates of neutropenia
NNRTIs and PIs in patients with HIV/HCV co- are also higher.
infection.
Co-administration with strong CYP3A4
inducers (including rifampin) is Simeprevir
Simeprevir is administered once daily in
contraindicated due to potential decrease in
combination with peginterferon and
serum levels of the anti-HCV agent, and co-
ribavirin for a total of 12 weeks in patients
administration with statin agents is
with compensated liver disease (including
contraindicated due to increased serum cirrhosis) that are infected with HCV
levels of the statin agent. genotype 1.
The effectiveness of hormonal Simeprevir must be taken with food to
contraceptives may be reduced by co- maximize absorption.
administration with boceprevir or telaprevir.
It is extensively bound to plasma proteins It is 59–76% bound to plasma proteins and
(> 99%), metabolized in the liver by CYP3A the effective half-life at steady state is 9–
pathways, and undergoes biliary excretion. 11 hours.
Its safety in patients with moderate to Telaprevir is metabolized by the CYP
severe liver insufficiency has not been pathways in the liver and is an inhibitor of
established. CYP3A4 and P-glycoprotein.
Mean simeprevir exposures are more than Co-administration of telaprevir with
threefold higher in Asian patients numerous drugs is contraindicated,
compared with Caucasians, leading to including rifampin, ergot derivatives,
potentially higher frequencies of adverse cisapride, lovastatin, simvastatin, alfuzosin,
events. sildenafil or tadalafil when used for
Simeprevir is a substrate and mild inhibitor pulmonary hypertension, pimozide, St.
of CYP3A and a substrate and inhibitor of John’s wort, triazolam, and midazolam.
P-gp and OATP1B1/3. The dosage of telaprevir must be increased
Co-administration with moderate or when coadministered with efavirenz, due
strong inhibitors or inducers of CYP3A may to lowered levels of telaprevir.
significantly increase or decrease the The most commonly reported adverse
plasma concentration of simeprevir. effects associated with telaprevir therapy
The presence of the NS3 Q80K are rash (30–55%), anemia, fatigue,
polymorphism at baseline is associated pruritus, nausea, and anorectal discomfort.
with reduced efficacy of therapy, and Severe rash or Stevens-Johnson syndrome
screening is recommended prior to the has been reported; in these patients, the
initiation of therapy. Emergence of amino drug should be stopped and not restarted.
acid substitutions resulting in decreased Rates of anemia are higher in patients
drug susceptibility has been documented taking telaprevir with peginterferon and
during therapy and may be associated with ribavirin than in those taking
reduced responsiveness. peginterferon and ribavirin alone (~ 36%
Reported adverse events include vs 17%, respectively).
photosensitivity reaction and rash (most Leukopenia, thrombocytopenia, increased
common within the first 4 weeks of serum bilirubin levels, hyperuricemia, and
therapy). anorectal burning may also occur.
Since simeprevir contains a sulfa moiety,
caution should be used in patients with a
history of sulfa allergy. RIBAVIRIN

Ribavirin is a guanosine analog that is


Telaprevir phosphorylated intracellularly by host cell
Therapy with telaprevir plus enzymes.
peginterferonn and ribavirin is Although its mechanism of action has not
administered for at least 12 weeks in been fully elucidated, it appears to interfere
treatment-naïve patients with HCV with the synthesis of guanosine triphosphate,
infection. to inhibit capping of viral messenger RNA,
As with boceprevir, the duration of therapy and to inhibit the viral RNA-dependent
is dependent on the achievement of polymerase of certain viruses.
undetectable virus. Ribavirin triphosphate inhibits the replication
Telaprevir must be taken with food to
of a wide range of DNA and RNA viruses,
maximize absorption.
including influenza A and B, parainfluenza,
respiratory syncytial virus, paramyxoviruses, The goal is to identify potent and well
HCV, and HIV-1. tolerated regimens that do not require
The absolute oral bioavailability of ribavirin is concurrent administration of interferon or
45–64%, increases with high-fat meals, and ribavirin; in addition agents are needed with
decreases with co-administration of antacids. activity against HCV genotypes other than 1
Plasma protein binding is negligible, volume (such as sofosbuvir).
of distribution is large, and cerebrospinal Other classes of agents in development
fluid levels are about 70% of those in plasma. include NS5A inhibitors (eg, daclatasvir), p7
Ribavirin elimination is primarily through the and NS4B inhibitors, cyclophilin inhibitors,
urine; therefore, clearance is decreased in and antisense oligonucleotides inhibiting
patients with creatinine clearances less than miR122 (eg, miravirsen).
30 mL/min.
ANTI-INFLUENZA AGENTS
Higher doses of ribavirin (ie, 1000–1200
mg/d, according to weight, rather than 800 Influenza virus strains are classified by their
mg/d) or a longer duration of therapy or core proteins (ie, A, B, or C), species of origin
both may be more efficacious in those with a (eg, avian, swine), and geographic site of
lower likelihood of response to therapy (eg, isolation.
those with genotype 1 or 4) or in those who Influenza A, the only strain that causes
have relapsed. pandemics, is classified into 16 H
This must be balanced with an increased (hemagglutinin) and 9 N (neuraminidase)
likelihood of toxicity. known subtypes based on surface proteins.
A dose-dependent hemolytic anemia occurs Although influenza B viruses usually infect
in 10–20% of patients. only people, influenza A viruses can infect a
Other potential adverse effects are variety of animal hosts.
depression, fatigue, irritability, rash, cough, Current influenza A subtypes that are
insomnia, nausea, and pruritus. circulating among worldwide populations
Contraindications to ribavirin therapy include include H1N1, H1N2, and H3N2. Fifteen
anemia, end-stage renal failure, ischemic subtypes are known to infect birds, providing
vascular disease, and pregnancy. an extensive reservoir.
Ribavirin is teratogenic and embryotoxic in Although avian influenza subtypes are
animals as well as mutagenic in mammalian typically highly species-specific, they have on
cells. rare occasions crossed the species barrier to
Patients exposed to the drug should not infect humans and cats.
conceive children for at least 6 months Viruses of the H5 and H7 subtypes (eg, H5N1,
thereafter. H7N7, and H7N3) may rapidly mutate within
poultry flocks from a low to high pathogenic
NEW & INVESTIGATIONAL AGENTS
form and have recently expanded their host
Second-generation NS3/NS4A protease range to cause both avian and human
inhibitors (eg, faldaprevir, simeprevir, disease.
asunaprevir), nucleoside/nucleotide NS5B Of particular concern is the avian H5N1 virus,
polymerase inhibitors (eg, sofosbuvir, see which first caused human infection (including
above), and non-nucleoside NS5B severe disease and death) in 1997 and has
polymerase inhibitors (eg, deleobuvir) are become endemic in Southeast Asian poultry
currently under clinical investigation. since 2003.
To date, the spread of H5N1 virus from Oral bioavailability is approximately 80%,
person to person has been rare, limited, and plasma protein binding is low, and
unsustained. concentrations in the middle ear and sinus
However, the emergence of the 2009 H1N1 fluid are similar to those in plasma.
influenza virus (previously called “swine flu”) The half-life of oseltamivir is 6–10 hours,
in 2009–2010 caused the first influenza and excretion is by glomerular filtration
and tubular secretion.
pandemic (ie, global outbreak of disease
Probenecid reduces renal clearance of
caused by a new flu virus) in more than 40
oseltamivir by 50%.
years.
Serum concentrations of oseltamivir
OSELTAMIVIR & ZANAMIVIR carboxylate, the active metabolite of
The neuraminidase inhibitors oseltamivir oseltamivir, increase with declining renal
and zanamivir, analogs of sialic acid, function; therefore, dosage should be
interfere with release of progeny influenza adjusted in patients with renal
virus from infected host cells, thus halting insufficiency.
the spread of infection within the Potential adverse effects include nausea,
respiratory tract. vomiting, and headache.
These agents competitively and reversibly Taking oseltamivir with food does not
interact with the active enzyme site to interfere with absorption and may
inhibit viral neuraminidase activity at low decrease nausea and vomiting.
nanomolar concentrations. Fatigue and diarrhea have also been
Inhibition of viral neuraminidase results in reported and appear to be more common
clumping of newly released influenza with prophylactic use.
virions to each other and to the membrane Rash is rare.
of the infected cell. Neuropsychiatric events (self-injury or
Unlike amantadine and rimantadine, delirium) have been reported, particularly
oseltamivir and zanamivir have activity in adolescents and adults living in Japan.
against both influenza A and influenza B Zanamivir is administered directly to the
viruses. respiratory tract via inhalation.
Early administration is crucial because Ten to twenty percent of the active
replication of influenza virus peaks at 24– compound reaches the lungs, and the
72 hours after the onset of illness. remainder is deposited in the oropharynx.
Initiation of a 5-day course of therapy The concentration of the drug in the
within 48 hours after the onset of illness respiratory tract is estimated to be more
decreases the duration of symptoms, viral than 1000 times the 50% inhibitory
shedding and transmission, and the rate of concentration for neuraminidase, and the
complications such as pneumonia, asthma, pulmonary half-life is 2.8 hours.
hospitalization, and mortality. Five to fifteen percent of the total dose (10
Once-daily prophylaxis is 70–90% effective mg twice daily for 5 days for treatment and
in preventing disease after exposure. 10 mg once daily for prevention) is
Oseltamivir is an orally administered absorbed and excreted in the urine with
prodrug that is activated by hepatic minimal metabolism.
esterases and widely distributed Potential adverse effects include cough,
throughout the body. bronchospasm (occasionally severe),
The dosage is 75 mg twice daily for 5 days reversible decrease in pulmonary function,
for treatment and 75 mg once daily for and transient nasal and throat discomfort.
prevention. Zanamivir administration is not
recommended for patients with
underlying airway disease.
Both oseltamivir and zanamivir are In the absence of resistance, both
available in intravenous formulations on a amantadine and rimantadine, at 100 mg
compassionate use basis from the twice daily or 200 mg once daily, are 70–
manufacturer. 90% protective in the prevention of clinical
Although resistance to oseltamivir and illness when initiated before exposure.
zanamivir may emerge during therapy and When begun within 1–2 days after the
be transmissible, nearly 100% of strains of onset of illness, the duration of fever and
H1N1, H3N2, and influenza B virus tested systemic symptoms is reduced by 1–2
by the Centers for Diseases Control for the days.
2012-2013 season retained susceptibility However, due to high rates of resistance in
to both agents. both H1N1 and H3N2 viruses, these agents
Oseltamivir resistance, however, has been are no longer recommended for the
documented in strains of the novel avian prevention or treatment of influenza.
H7N9 virus, in one instance appearing to The most common adverse effects are
emerge during treatment. gastrointestinal (nausea, anorexia) and
central nervous system (nervousness,
difficulty in concentrating, insomnia, light-
AMANTADINE & RIMANTADINE headedness).
Amantadine (1-aminoadamantane More serious side effects (eg, marked
hydrochloride) and its α-methyl derivative, behavioral changes, delirium,
rimantadine, are tricyclic amines of the hallucinations, agitation, and seizures) may
adamantane family that block the M2 be due to alteration of dopamine
proton ion channel of the virus particle and neurotransmission (see Chapter 28); are
inhibit uncoating of the viral RNA within less frequent with rimantadine than with
infected host cells, thus preventing its amantadine; are associated with high
replication. plasma concentrations; may occur more
They are active against influenza A only. frequently in patients with renal
Rimantadine is four to ten times more insufficiency, seizure disorders, or
active than amantadine in vitro. advanced age; and may increase with
Amantadine is well absorbed and 67% concomitant antihistamines,
protein-bound. Its plasma half-life is 12–18 anticholinergic drugs,
hours and varies by creatinine clearance. hydrochlorothiazide, and trimethoprim-
Rimantadine is about 40% protein-bound sulfamethoxazole.
and has a half-life of 24–36 hours. Clinical manifestations of anticholinergic
Nasal secretion and salivary levels activity tend to be present in acute
approximate those in the serum, and amantadine overdose.
cerebrospinal fluid levels are 52–96% of Both agents are teratogenic and
those in the serum; nasal mucus embryotoxic in rodents, and birth defects
concentrations of rimantadine average have been reported after exposure during
50% higher than those in plasma. pregnancy.
Amantadine is excreted unchanged in the
urine, whereas rimantadine undergoes
extensive metabolism by hydroxylation, INVESTIGATIONAL AGENTS
conjugation, and glucuronidation before
The neuraminidase inhibitor peramivir, a
urinary excretion.
Dose reductions are required for both cyclopentane analog, has activity against
agents in the elderly and in patients with both influenza A and B viruses.
renal insufficiency, and for rimantadine in Peramivir received temporary emergency use
patients with marked hepatic insufficiency. authorization by FDA for intravenous
administration in November 2009 due to the Intravenous ribavirin decreases mortality
H1N1 pandemic, but is not now approved for in patients with Lassa fever and other viral
use in the USA. hemorrhagic fevers if started early.
Reported side effects include diarrhea, High concentrations inhibit West Nile virus
nausea, vomiting, and neutropenia. in vitro, but clinical data are lacking.
A long-acting neuraminidase inhibitor, Clinical benefit has been reported in cases
of severe measles pneumonitis and certain
laninamivir octanoate, may retain activity
encephalitides, and continuous infusion of
against oseltamivir-resistant virus.
ribavirin has decreased virus shedding in
DAS181 is a host-directed antiviral agent that
several patients with severe lower
acts by removing the virus receptor, sialic respiratory tract influenza or parainfluenza
acid, from adjacent glycan structures. infections.
At steady state, cerebrospinal fluid levels
OTHER ANTIVIRAL AGENTS
are about 70% of those in plasma.

INTERFERONS
Interferons have been studied for PALIVIZUMAB
numerous clinical indications. In addition Palivizumab is a humanized monoclonal
to HBV and HCV infections (see antibody directed against an epitope in
Antihepatitis Agents), intralesional the A antigen site on the F surface protein
injection of interferon alfa-2b or alfa-n3 of RSV.
may be used for treatment of condylomata It is licensed for the prevention of RSV
acuminata (see Chapter 61). infection in high-risk infants and children,
such as premature infants and those with
bronchopulmonary dysplasia or
RIBAVIRIN
congenital heart disease.
In addition to oral administration for HCV
A placebo-controlled trial using once-
infection in combination with interferon
monthly intramuscular injections (15
alfa (see Antihepatitis Agents), aerosolized
mg/kg) for 5 months beginning at the start
ribavirin is administered by nebulizer (20
of the RSV season demonstrated a 55%
mg/mL for 12–18 hours per day) to
reduction in the risk of hospitalization for
children and infants with severe
RSV in treated patients, as well as
respiratory syncytial virus (RSV)
decreases in the need for supplemental
bronchiolitis or pneumonia to reduce the
oxygen, the illness severity score, and the
severity and duration of illness.
need for intensive care.
Aerosolized ribavirin has also been used to
Although resistant strains have been
treat influenza A and B infections but has
isolated in the laboratory, no resistant
not gained widespread use.
clinical isolates have yet been identified.
Systemic absorption is low (< 1%).
Potential adverse effects include upper
Aerosolized ribavirin may cause
respiratory tract infection, fever, rhinitis,
conjunctival or bronchial irritation and the
rash, diarrhea, vomiting, cough, otitis
aerosolized drug may precipitate on
media, and elevation in serum
contact lenses.
aminotransferase levels.
Ribavirin is teratogenic and embryotoxic.
Agents under investigation for the
Health care workers and pregnant women
treatment or prophylaxis of patients with
should be protected against extended
RSV infection include the RNA interference
inhalation exposure.
(RNAi) therapeutic ALN-RSV01and the
benzodiazepine RSV604.
IMIQUIMOD
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