Sunteți pe pagina 1din 17

Drugs 2009; 69 (8): 1059-1075

ADIS DRUG EVALUATION 0012-6667/09/0008-1059/$55.55/0

ª 2009 Adis Data Information BV. All rights reserved.

Raltegravir
A Review of its Use in the Management of HIV Infection in
Treatment-Experienced Patients
Jamie D. Croxtall and Susan J. Keam
Wolters Kluwer Health | Adis, Auckland, New Zealand, an editorial office of Wolters Kluwer
Health, Philadelphia, Pennsylvania, USA

Various sections of the manuscript reviewed by:


F. Cainelli, School of Medicine, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana;
B. Gazzard, St Stephen’s Centre, Chelsea and Westminster Hospital, London, UK; D.T. Jayaweera, Division of
Infectious Diseases, University of Miami School of Medicine, Miami, Florida, USA; M. Nelson, HIV Medicine,
Chelsea and Westminster Hospital, London, UK; R. Wood, Institute of Infectious Disease and Molecular
Medicine, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.

Data Selection
Sources: Medical literature published in any language since 1980 on ‘raltegravir’, identified using MEDLINE and EMBASE, supplemented
by AdisBase (a proprietary database of Wolters Kluwer Health | Adis). Additional references were identified from the reference lists of
published articles. Bibliographical information, including contributory unpublished data, was also requested from the company developing
the drug.
Search strategy: MEDLINE, EMBASE and AdisBase search term was ‘raltegravir’. Searches were last updated 20 May 2009.
Selection: Studies in patients with HIV infection who received raltegravir. Inclusion of studies was based mainly on the methods section of
the trials. When available, large, well controlled trials with appropriate statistical methodology were preferred. Relevant pharmacodynamic
and pharmacokinetic data are also included.
Index terms: Raltegravir, HIV infection, antiretroviral therapy, integrase inhibitor, pharmacodynamics, pharmacokinetics, therapeutic use,
tolerability.

Contents
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1060
1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061
2. Pharmacodynamic Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061
2.1 Mechanism of Action. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061
2.2 Antiretroviral Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1062
2.3 Resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1062
2.4 Other Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1063
3. Pharmacokinetic Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1063
3.1 Absorption and Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1063
3.2 Metabolism and Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064
3.3 Special Populations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064
3.4 Drug Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1065
4. Therapeutic Efficacy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1066
5. Tolerability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1069
6. Dosage and Administration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1070
7. Place of Raltegravir in the Management of HIV Infection in Treatment-Experienced Patients. . . . . 1070
1060 Croxtall & Keam

Summary
Abstract Raltegravir (Isentress), an integrase inhibitor, inhibits the insertion of HIV-1
complementary DNA into the host genome. It is indicated in combination with
other antiretroviral therapy (ART) agents for the treatment of HIV-1 infection
in treatment-experienced adult patients who have evidence of viral replication
and HIV-1 strains resistant to multiple ART agents. It is the first of a new class
of ART agents to be approved that, as a result of a different mechanism of
action to other ART agents, has good activity against multidrug-resistant HIV-1
strains.

In clinical trials in treatment-experienced patients with HIV-1 infection and


evidence of viral replication, the addition of oral raltegravir to an optimized
background therapy (OBT) regimen improved virological and immunological
responses at 16 and 48 weeks to a greater extent than placebo plus OBT. Ralte-
gravir therapy was generally well tolerated, with a similar incidence of mild to
moderate adverse events in the treatment and placebo arms. The introduction of
integrase inhibitors extends the options available for managing treatment-
experienced patients with multiple-drug-resistant HIV-1 infection. Results to date
suggest that the combination of raltegravir and OBT will be a valuable treatment
option for this difficult-to-treat patient group.

Pharmacological Raltegravir is a selective inhibitor of integrase, an HIV-1-specific enzyme that


Properties is responsible for the insertion of viral complimentary DNA into the host
genome. In in vitro studies, the 95% inhibitory concentration (IC95) of raltegravir
in human T-lymphoid cells infected with a laboratory strain of HIV-1 was
31 nmol/L and ranged from 6 to 50 nmol/L in human peripheral blood mono-
nuclear cells, infected with clinical isolates of HIV-1, including strains resistant
to other classes of ART. Specific mutations within the HIV-1 integrase gene
(predominantly at Q148, N155 or Y143) are correlated with virological resistance
to raltegravir. Patients with higher baseline levels of HIV-1 RNA and less active
OBT regimens are at the highest risk of integrase mutations arising during
treatment.

Oral raltegravir is rapidly absorbed, with a mean maximum plasma con-


centration reached at a median time of »1 hour. The mean plasma concentration
of raltegravir at the end of a 12-hour dose administration interval after a 400 mg
dose exceeded the IC95 against HIV-1 in vitro. Raltegravir is metabolized in hu-
mans through uridine diphosphate-glucuronosyltransferase-mediated glucur-
onidation in the liver, and is eliminated in faeces and urine. The mean elimination
half-life of raltegravir is 7–12 hours. Raltegravir does not significantly affect the
activity of the cytochrome P450 system and therefore is not expected to interact
with other ART agents that are substrates for these enzymes.

Therapeutic Efficacy In the two phase III BENCHMRK trials (n = 699), orally administered raltegravir
400 mg twice daily plus OBT improved both virological and immunological out-
comes in treatment-experienced patients with HIV-1 infection, evidence of viral
replication and HIV-1 strains resistant to multiple ART agents. At 16 weeks in the
individual trials and at 48 weeks in the combined analysis, raltegravir plus
OBT reduced HIV-1 RNA levels to <400 copies/mL (primary endpoint) and

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1061

<50 copies/mL in a significantly greater number of patients and significantly


increased CD4+ cell counts from baseline when compared with placebo plus OBT.
Moreover, prespecified subgroup analyses indicated that in the BENCHMRK
trials, the greater efficacy of raltegravir plus OBT compared with placebo plus
OBT was maintained in patients with baseline characteristics that typically pre-
dict a poor response to ART therapy, for example, higher HIV-1 RNA levels,
lower CD4+ cell counts and less active OBT regimens.

Tolerability Raltegravir was generally well tolerated when used in combination with OBT
regimens in treatment-experienced patients with HIV-1 infection in trials of up to
48 weeks’ duration. The majority of adverse events were mild to moderate in
severity, and of similar incidence in the raltegravir plus OBT and placebo plus
OBT treatment arms; treatment-related discontinuations were uncommon. The
most common drug-related adverse events reported were diarrhoea, headache,
nausea and fatigue.

1. Introduction Integrase is an HIV-1-specific enzyme essential


for viral replication. It catalyses the insertion of
The 2008 Joint United Nations Programme on viral complementary DNA (cDNA) into the
HIV/AIDS report on the global AIDS epidemic genome of host cells.[7] Inhibition of this novel
estimates that in 2007 there were 33 million peo- target may be of value in patients with HIV-1
ple infected with HIV type-1 (HIV-1) world- variants that exhibit resistance to conventional
wide.[1] There were 2.7 million new infections ART agents.
occurring in 2007, which is lower than in previous Raltegravir (Isentress) is the first ART in-
years and, as a consequence, the epidemic ap- tegrase inhibitor to be approved for use in the
pears to be stabilizing;[1] however, the prevalence treatment of HIV-1 infection. This article reviews
of infection remains unacceptably high.[1-3] the pharmacological properties of raltegravir,
Although antiretroviral therapy (ART) regi- and its clinical efficacy and tolerability profile in
mens have significantly reduced the morbidity and treatment-experienced patients with HIV-1 infec-
mortality associated with HIV-1 infections, there is tions resistant to other ART agents.
currently no cure for the disease. Moreover, for all
ART regimens, the emergence of both evolved and 2. Pharmacodynamic Properties
transmitted resistance presents a significant com-
promise for effective long-term treatment suc- The pharmacodynamic properties of raltegravir
cess.[2,4,5] Indeed, estimates suggest that as many as have been reviewed previously in detail.[7-10] There-
76% of treatment-experienced patients who are fore, this section provides an overview of these and
viraemic harbour viruses that exhibit resistance,[6] more recently published data,[11-17] supplemented,
with a prevalence of three-class drug resistance of where appropriate, by the manufacturer’s pre-
more than 13% in viraemic treatment-experienced scribing information.[18,19]
patients in the US.[6]
Treatment successes cannot be sustained with- 2.1 Mechanism of Action
out reducing the incidence of new infections,
reducing drug toxicities and tackling the problem Raltegravir is a hydroxypyrimidinone carbox-
of ART resistance.[2,4,5] Therefore, there remains amide that shares the b-hydroxy ketone struc-
an ongoing need for the development of new tural motif of the diketo acid class of compounds
treatment modalities for patients with HIV-1 (figure 1). This motif is a defining moiety of the
infection. integrase inhibitor class. Raltegravir prevents

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1062 Croxtall & Keam

O protease inhibitors (PIs) and a fusion inhibitor,


N H3C O–K+ F against human T-lymphoid cells infected with
N N
HIV-1 variant H9IIIB.[18]
H3C H H
O N
N
N HIV-2 exhibits inherent resistance to NNRTIs
and fusion inhibitors, and is less sensitive than
O H3C CH3 O
HIV-1 to several PIs in vitro.[23] However, ralte-
Fig. 1. Chemical structure of raltegravir. gravir showed good in vitro inhibitory activity
against 14 clinical isolates of HIV-2 (median IC50
covalent insertion of viral HIV-1 cDNA into the and IC95 values of 2.4 and 12.5 nmol/L) in human
host cell genome.[9-11,20-22] Importantly, host cells PBMC assays[13] and in an isolate of HIV-2 in
do not express integrase activity; thus, raltegravir CEMx174 cells (T-cell/B-cell hybrid cell line)
is unlikely to affect normal cell activity.[7-9] [IC95 6 nmol/L].[18]
Like other integrase inhibitors, raltegravir
selectively blocks the strand-transfer activity of 2.3 Resistance
integrase at nanomolar concentrations (appar-
ent 50% inhibitory concentration [IC50] of Virological failure of raltegravir in patients
2–7 nmol/L).[11,22] Integrase inhibition prevents with HIV-1 infection appears to be correlated
the insertion of linear HIV-1 cDNA into the host with specific mutations at several key sites
cell genome. Consequently, the formation of HIV-1 within the integrase gene.[18,19,24] Several stu-
provirus, which is necessary for the production of dies[14-16,18,19,24] have indicated that mutations
progeny virus and infection of new cells, is that result in amino acid substitutions at one of
prevented.[7-9] By contrast, other human phos- three signature sites, including Q148 (changed to
phoryltransferases, such as the polymerase and H, K or R; i.e. Q148H/K/R), N155H or
ribonuclease H activities of HIV-1 reverse tran- Y143C/H/R, together with one or more addi-
scriptase and DNA polymerase a, b and g, are not tional substitutions (L74I/M/R, E92Q, T97A,
significantly inhibited by raltegravir in vitro.[9] E138A/K, G140A/S, V151I, G163R, H183P,
Y226D/F/H, S230R, D232N, T66A and
2.2 Antiretroviral Activity
E157Q[15,16,18,19,24]), play a crucial role in the
development of resistance to raltegravir. A small
Raltegravir is a potent inhibitor of viral repli- study (n = 9) in treatment-experienced patients
cation in vitro, including ART-resistant strains.[18,19] receiving salvage therapy (which included ralte-
In studies in human T-lymphoid cell cultures in- gravir) who subsequently developed virological
fected with a laboratory strain of HIV-1 (variant failure to raltegravir showed that the E92Q,
H9IIIB), the 95% inhibitory concentration (IC95) G140S and Q148H substitutions of HIV-1 in-
of raltegravir was 31 nmol/L.[18,19] In mitogen- tegrase were associated with a 7- to 8-fold
activated human peripheral blood mononuclear decrease in susceptibility to raltegravir in vitro,
cells (PBMCs) infected with a variety of HIV-1 and the N155H substitution was associated with
clinical isolates, including strains resistant to a 14-fold decrease.[16]
ARTs of other classes, raltegravir IC95 values In the ongoing phase III BENCHMRK
ranged from 6 to 50 nmol/L.[18] Mean IC95 values (Blocking integrase in treatment Experienced
did not differ between B subtype and non-B patients with a Novel Compound against HIV:
subtype HIV-1 clinical isolates assessed in human MeRcK) trials (see section 4 for trial design de-
PBMC replication assays (14 and 21 nmol/L).[12] tails), 105 of 462 raltegravir recipients (23%) ex-
Additive-to-synergistic activity was evident hibited virological failure at an interim assess-
in vitro when raltegravir was used in combination ment time of 48 weeks.[14] Genotyping of HIV-1
with other ART agents, including non-nucleoside integrase in 94 of these patients showed that 64
reverse transcriptase inhibitors (NNRTIs), nu- had a mutation known to confer raltegravir re-
cleoside reverse transcriptase inhibitors (NRTIs), sistance, with 54 having amino acid substitutions

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1063

at either Q148, N155 or Y143.[14] Of the remain- manufacturer’s prescribing information[18,19] is


ing 30 patients, 25 had no amino acid changes also included.
from baseline sequence and five had changes of
unknown phenotypic effect.[14] In addition, most 3.1 Absorption and Distribution
(48 of 64) patients with resistance had two or
more mutations associated with resistance. Sub- The absolute bioavailability of raltegravir has
group analyses of the combined BENCHMRK not been established. The pharmacokinetic para-
trials (n = 699) showed that patients with higher meters of orally administered raltegravir (400 mg
baseline HIV-1 RNA levels or genotypic or phe- single dose or 400 mg twice daily for 10 days) in
notypic scores for optimized background therapy healthy volunteers and treatment-naive patients
(OBT) of 0 had an increased risk of HIV-1 in- with HIV-1 infection are summarized in table I.
tegrase mutations arising during treatment.[14] In a single-dose escalation study in fasted healthy
On the other hand, clade-specific polymorph- male volunteers, the mean maximum plasma con-
isms of the HIV-1 integrase gene do not appear to centration (Cmax), the mean plasma concentra-
contribute to reduced susceptibility to raltegravir.[17] tion at 12 hours post-dose (C12) and the mean
The emergence of mutations Q148K/R and area under the plasma concentration-time curve
N155H have been identified in the integrase gene (AUC) from time zero to infinity (AUC¥) of or-
of HIV-2 infected patients with raltegravir re- ally administered raltegravir increased approxi-
sistance,[13,25] suggesting a common pathway to mately proportionally with doses of 10–1200 mg,
integrase inhibitor resistance with HIV-1. and slightly less than proportionally in doses
up to 1600 mg.[30] Absorption was rapid, with
a median time to Cmax (tmax) of approximately
2.4 Other Effects 1 hour.[30] Systemic exposure to single-dose ral-
Oral raltegravir 400 mg twice daily for tegravir 400 mg was similar in fasted healthy male
48 weeks had little effect on fasting serum lipids and female volunteers, as assessed by the
in the BENCHMRK trials, as evidenced by non- AUC¥.[30] The rate, but not the extent, of ralte-
significant changes relative to placebo in total gravir absorption was slowed when administered
cholesterol (increased by 7.4%) and low-density with a high-fat meal in healthy volunteers.[29]
lipoprotein (LDL) cholesterol (increased by These effects, and the associated small increase
6.2%).[26] Unlike many other ART agents, ralte- in systemic exposure, were not considered to
gravir does not appear to be associated with the be clinically meaningful, and raltegravir can be
development of clinically relevant dyslipidaemia administered with or without food.[18,19]
or hypercholesterolaemia.[19] Following multiple-dose administration of oral
Furthermore, a single supratherapeutic oral raltegravir 100–800 mg twice daily for 10 days in
dose (1600 mg) of raltegravir did not prolong the healthy male volunteers, both the AUC during a
corrected QT (Fridericia method) [QTcF] inter- dose administration interval (0–12 hours) and the
val in healthy volunteers in a thorough QT/QTc Cmax increased approximately proportionally with
study (n = 31).[27] dose, and steady-state raltegravir concentrations
were achieved within 2 days.[30] C12 values in-
creased slightly less than proportionally with
3. Pharmacokinetic Properties dose;[30] however, at all dosages the C12 values for
raltegravir were higher than 33 nmol/L, thus ex-
This section reviews the pharmacokinetic ceeding the raltegravir IC95 against most strains
properties of raltegravir in healthy volunteers of HIV-1 in vitro (section 2.2).
(n = 6–57)[28-35] and treatment-naive patients with In treatment-naive patients with HIV-1 infec-
HIV-1 infection (n = 6),[36] focusing, where possi- tion, Cmax, C12 and AUC¥ values increased pro-
ble, on data using the approved dosage of 400 mg portionally with raltegravir 100–400 mg twice
twice daily. Additional information from the daily after 10 days of treatment, with no further

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1064 Croxtall & Keam

Table I. Mean pharmacokinetic values for single- (400 mg)[30] or multiple-dose (400 mg twice daily for 10 days)[30,36] orally administered
raltegravir as monotherapy in randomized, double-blind, placebo-controlled trials in healthy volunteers[30] and treatment-naive patients with
HIV-1 infection[36]
Parameter Healthy male volunteers[30]a Treatment-naive patients[36]
single dose (n = 6)b multiple dose (n = 8)c multiple dose (n = 6)c
Cmax (mmol/L) 10.6 11.2 4.5
tmax (h) 1.0d 1.0d 1.7


C12 (nmol/L) 81.3 200.6 142
AUC (mmol h/L) 24.6e 28.7f 14.2f
t1/2a (h) 1.1 1.1
t1/2b (h) 6.9 10.7
CLR (L/h) 3.6 3.6
a Lactose formulation of raltegravir.
b Fasted state.
c Steady-state values at day 10.
d Median value.
e From time zero to infinity.
f From 0 to 12 hours.
AUC = area under the plasma concentration-time curve; C12 = plasma concentration at the end of the dose administration interval (12 h);
CLR = renal clearance; Cmax = maximum plasma concentration; tmax = time to reach Cmax; t1/2 = elimination half-life of the initial (a) or terminal (b)
phases.

increase for the 600 mg dosage.[36] Absorption undetectable after 24 hours.[31] The appa-
was rapid, with a mean tmax of 1.3–2 hours. Mean rent mean terminal elimination half-life after
C12 values at all raltegravir dosages at day 10 single or repeated administration of raltegravir
exceeded 33 nmol/L.[36] 10–1600 mg was 6–12 hours, but there is also a
Approximately 83% of the drug is bound to shorter half-life initial phase (»1 hour) which
human plasma protein over the concentration accounts for most of the AUC.[18,19,30]
range 2–10 mmol/L.[18] The renal clearance rate of raltegravir was
3.6 L/h for healthy volunteers receiving the recom-
3.2 Metabolism and Elimination mended dosage of 400 mg twice daily (table I).[30]
Raltegravir was eliminated in the faeces (51%)
Raltegravir is metabolized in humans pri- and urine (32%) of healthy volunteers following
marily through uridine diphosphate-glucuronosyl- oral administration of the 14C-labelled drug.[31]
transferase (UGT)-mediated glucuronidation in The glucuronide metabolite of raltegravir ac-
the liver, predominantly via UGT1A1.[31] Ap- counted for 23% of the label recovered in urine,
proximately 33% of raltegravir recovered in the whereas only the parent compound was found in
plasma of healthy volunteers was found to be the the faecal fraction.[31]
glucuronide derivative, with the remainder ac-
counted for by the parent compound.[31] There 3.3 Special Populations
was no evidence for oxidative metabolism of
raltegravir in isolated human liver microsomes Age, sex, race, moderate hepatic impairment
and the drug did not interact with cytochrome and severe renal impairment had no appreciable
P450 (CYP).[31] effect on the pharmacokinetic profile of ralte-
Plasma concentrations of orally administered gravir, and dosage adjustments are not necessary
raltegravir declined rapidly in healthy volun- in these patient groups.[18,19,37] However, the
teers following a single 200 mg dose and were pharmacokinetics of raltegravir in children and in

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1065

patients with severe hepatic impairment have not administered agents and raltegravir in healthy
been determined. The extent to which raltegravir is volunteers, and these are summarized in table
dialyzable is unknown; therefore, administration II,[32-35,38,39] together with recommendations
prior to a dialysis session is not recommended.[18,19] for dosage adjustment for patients with HIV-1
Following a single 400 mg dose, plasma con- infection.[18,19] There are no drug interaction
centrations of raltegravir were slightly higher for studies of raltegravir with darunavir.[40]
individuals with the UGT1A1*28/*28 genotype Raltegravir does not significantly affect the ac-
compared with the normal UGT1A1*1/*1 geno- tivity of the CYP isoenzymes, including CYP3A4,
type, when matched for age, sex, race and body nor does it inhibit UGT1A1 and UGT2B7 activity
mass index.[28] The geometric mean ratio and P-glycoprotein-mediated transport in vitro.[18,19]
(UGT1A1*28/*28 : UGT1A1*1/*1) for AUC¥ was Therefore, raltegravir is not expected to interact
1.41, for Cmax was 1.40 and for C12 was 1.91; with agents that are substrates of these enzymes,
however, tmax and elimination half-life values were including PIs, NNRTIs, methadone, opioid an-
similar for both genotypes.[28] These differences algesics, statins, azole antifungals, oral contracep-
are not considered to be clinically significant and tives, proton pump inhibitors and anti-erectile
dosage adjustments are not required in individuals dysfunction agents.[18,19]
with the UGT1A1*28/*28 polymorphism.[18,19] This is largely confirmed in the studies pre-
sented in table II. However, coadministration
3.4 Drug Interactions
with the proton pump inhibitor omeprazole re-
sulted in an increase in plasma levels of ralte-
Several studies have investigated the pharma- gravir.[35] As a consequence, coadministration of
cokinetic interactions between commonly co- raltegravir with agents that increase gastric pH is

Table II. Pharmacokinetic interactions between raltegravir (RAL) and other coadministered drugs in healthy subjects and recommended
dosage adjustments. The manufacturer’s prescribing information[18,19] should be consulted for further information on drug interactions
Dosage regimen (dose in mg) [frequency] No. of Ratio with : without coadministered drug Recommended dosage
Coadministered drug RAL subjects Cmax AUC C12 adjustment[18,19]
Effect of coadministered drug on the mean pharmacokinetic parameters of RAL
ATA 300 + RIT 100 [od][32] 400 [bid] 10 1.24 1.41a 1.77 None required
EFA 600 [od][33] 400 [sd] 9 0.64 0.64b 0.79 None required
OME 20 [sd][35] 400 [sd] NR 4.15 3.12b 1.46 Coadministration not
recommended
RIF 600 [od][38] 400 [sd] 9 0.62 0.60b 0.39 Caution required
RIT 100 [bid][33] 400 [sd] 10 0.76 0.84b 0.99 None required
TEN 300 [od][18] 400 [bid] 9 1.64 1.49c 1.03 None required
TIP 500 + RIT 200 [bid][39] 400 [bid] 18 0.82 0.76b 0.45 None required
Effect of RAL on the mean pharmacokinetic parameters of coadministered drug
ETH [NR][19] 400 [bid] NR 1.01 0.98c NR None required
MID 2 [sd] [34]
400 [bid] 10 1.03 0.92b NR None required
NOR [NR][19] 400 [bid] NR 1.29 1.14c NR None required
TEN 300 [od][19] 400 [bid] 9 0.67 0.9c 0.87 None required
a From 0 to 12 hours.
b From 0 to ¥.
c Interval not defined.
ATA = atazanavir; AUC = area under the plasma concentration-time curve; bid = twice daily; C12 = plasma concentration at the end of the
dose administration interval (12 h); Cmax = maximum plasma concentration; EFA = efavirenz; ETH = ethinyl estradiol; MID = midazolam;
NOR = norelgestromin; NR = not reported; od = once daily OME = omeprazole; RIF = rifampicin; RIT = ritonavir; sd = single dose;
TEN = tenofovir; TIP = tipranavir.

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1066 Croxtall & Keam

not recommended unless this is unavoidable.[19] a raltegravir 400 mg twice daily plus etravirine and
Additionally, because raltegravir is metabolized darunavir/ritonavir regimen, and is not discussed
by UGT-mediated glucuronidation, agents such further. Furthermore, a small (n = 35) switching
as rifampicin (rifampin), which are strong in- trial suggested that all patients not tolerating en-
ducers of UGT enzymes, should be coadminis- fuvirtide could replace enfuvirtide with raltegravir
tered with caution and modification in raltegravir in the combined ART regimen without loss of
dosage may be required (section 6).[18,19] viral suppression for up to a median time of
7 months (except one patient with a detectable, but
4. Therapeutic Efficacy transient, viral load after 5 months).[46] Similarly,
a randomized, noninferiority, switching trial in
The therapeutic efficacy of raltegravir in com- highly treatment-experienced patients (n = 170)
bination with other antiretroviral agents has been indicated that raltegravir plus OBT was non-
evaluated in patients with HIV-1 infection who are inferior to enfuvirtide plus OBT in terms of viral
treatment-naive[36,41,42] or treatment-experienced suppression.[47] These trials are also not discussed
with multidrug ART resistance.[14,26,43] However, further.
raltegravir is not approved for use in treatment- Patients presented with similar baseline char-
naive patients and this section reviews clinical trial acteristics in both BENCHMRK trials: with a
data in treatment-experienced patients only. median age range of 43–47 years, median plasma
Clinical data reviewed in this section in pa- HIV-1 RNA levels of 4.6–4.8 log10 copies/mL,
tients (aged ‡16 years) with ART-resistant HIV-1 median CD4+ cell counts of 102–140 cells/mm3,
receiving oral raltegravir 400 mg twice daily (the median duration of prior ART use of 10–11 years,
recommended dosage; see section 6) are primarily median number of ART drugs in OBT of four, a
derived from two BENCHMRK trials.[14,26] history of AIDS in 89–93.5%, a co-infection of
BENCHMRK-1 and BENCHMRK-2 (MK-0518 hepatitis B and/or C in 7.6–22.9% and a geno-
protocols 018 and 019) are identically designed, typic or phenotypic score of 0 in 10.4–30.2%;
ongoing (scheduled to run at least 156 weeks), 84.1–91.3% were male.[26] Randomization of pa-
randomized, double-blind, placebo-controlled, tients to receive either raltegravir 400 mg twice
multicentre, phase III trials in ART-experienced daily plus OBT (n = 232 or 230) or placebo twice
patients with virological failure (HIV-1 RNA daily plus OBT (n = 118 or 119) was stratified
levels >1000 copies/mL) when receiving ART ther- according to the extent of resistance to prior PI
apy, and documented phenotypic or genotypic treatment and the inclusion of enfuvirtide in the
resistance to at least one drug in each of three OBT.[26] In addition to prior ART treatment,
approved classes of oral ART drugs (NRTIs, both genotypic and phenotypic resistance testing
NNRTIs, PIs).[14,26] BENCHMRK-1 is being of patients determined the selection of OBT be-
conducted in Europe, Asia/Pacific and Peru, fore randomization.[26]
whereas BENCHMRK-2 is being conducted in Patients with renal insufficiency (serum crea-
North, Central and South America.[14,26] tinine levels more than 2 · the upper limit of
The BENCHMRK clinical trial data are sup- normal [ULN]) or acute or decompensated
ported by a phase II, randomized, double-blind, chronic hepatitis were excluded from the study.
dose-finding trial in ART-experienced patients Patients with stable chronic hepatitis B or hepa-
with HIV-1 infection (protocol 005)[44] that esta- titis C were permitted if their serum aminotrans-
blished raltegravir 400 mg twice daily as the ferase levels were <5 · the ULN, and patients
optimum dosage regimen. This study has been with active cancer were enrolled if they did not
reviewed previously in detail[9] and is not dis- require chemotherapy and the cancer was un-
cussed further. A noncomparative, open-label, likely to affect the study outcome.[26]
multicentre, ongoing, phase II French study In both BENCHMRK studies, the prespeci-
(ANRS 139 TRIO)[45] in patients with ART- fied primary outcome measure was defined as the
resistant HIV-1 (n = 103) indicated the efficacy of proportion of patients achieving suppression of

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1067

HIV-1 RNA levels to <400 copies/mL after 16 achieved this endpoint, compared with just over
weeks of treatment.[26] Prespecified secondary 40% of those receiving placebo plus OBT (both
outcome measures were defined as the proportion p < 0.001). Furthermore, HIV-1 RNA levels were
of patients with HIV-1 RNA levels <50 suppressed to <50 copies/mL in a significantly
copies/mL and the change from baseline in CD4+ greater number of raltegravir plus OBT than in
cell counts at weeks 16 and 48.[26] A combined placebo plus OBT recipients (table III).[26] These
analysis of both BENCHMRK trials was also results were confirmed when the more stringent
prespecified to provide precise estimates of viro- criterion of patient noncompletion counted as
logical response rates. Logistic regression analy- treatment failure was used in the individual stu-
sis showed that as a result of the consistency of dies and the combined analysis (78% vs 42% for
efficacy between treatment groups, exploratory patients achieving HIV-1 RNA levels <400
subgroup analyses of pooled data were fea- copies/mL and 62% vs 35% for patients achieving
sible.[14,26] HIV-1 RNA levels <50 copies/mL).[26]
All treated patients were included in the final The greater efficacy of raltegravir plus OBT
efficacy analysis. Missing data were analysed in over placebo plus OBT in the BENCHMRK
the following three ways: the primary efficacy trials was maintained following 48 weeks’ treat-
analysis counted treatment-related discontinua- ment (table III).[26] At least 72% of raltegravir
tions as treatment failures at subsequent time- plus OBT recipients in the combined analysis had
points; a second, worst-case approach counted plasma HIV-1 RNA levels of <400 copies/mL and
noncompletion for any reason as treatment fail- 62% achieved levels of <50 copies/mL. By con-
ure; and a third, observed-failure approach trast, only 37% and 33% of those receiving pla-
counted discontinuation due to a lack of efficacy cebo plus OBT achieved these endpoints.[26]
as treatment failure.[26] Moreover, the proportion of patients with HIV-1
Prespecified subgroup analyses of combined RNA levels of <50 copies/mL for recipients of
data from the two BENCHMRK trials were raltegravir plus OBT or placebo plus OBT was
conducted at week 48 according to demographic little changed between week 16 using treatment-
characteristics, baseline HIV-1 RNA levels, related discontinuation as virological failure data
CD4+ counts, the inclusion of active PIs in the (62–63% vs 33–36%) and week 48 using patient
OBT and genotypic/phenotypic testing for HIV-1 noncompletion as virological failure data
resistance; however, the analysis of enfuvirtide (60–65% vs 31–35%) [table III].[26]
plus darunavir in OBT was specified after CD4+ cell counts rose from baseline (102–140
unblinding.[14] cells/mm3) by a mean of 83 cells/mm3 in
Orally administered raltegravir 400 mg twice BENCHMRK 1 and 86 cells/mm3 in BENCHMRK
daily plus OBT significantly improved virological 2 following 16 weeks’ treatment with raltegravir
and immunological outcomes in treatment- plus OBT compared with 31 and 40 cells/mm3
experienced patients with HIV-1 infection and for recipients of placebo plus OBT (values esti-
triple class resistance compared with placebo mated from a graph; significance not reported)
twice daily plus OBT in the BENCHMRK [table III].[26] The increase from baseline in CD4+
trials.[26] cell counts was maintained through to 48 weeks in
The proportion of patients achieving HIV-1 both BENCHMRK trials. In the combined analy-
RNA levels of <400 copies/mL, with treatment- sis using observed lack of efficacy as virological
related discontinuations counted as virological failure data, CD4+ cell counts increased from
failure, was significantly greater in the raltegravir baseline by a mean of 109 cells/mm3 for recipients
plus OBT treatment group than in the placebo of raltegravir plus OBT versus 45 cells/mm3 for
plus OBT treatment group in both those receiving placebo plus OBT (p < 0.001) at
BENCHMRK-1 and BENCHMRK-2 trials at week 48 (table III).[26]
week 16 (primary endpoint; table III).[26] In both Using an observed lack of efficacy as treat-
trials, 78% of raltegravir plus OBT recipients ment failure analysis, the mean change from

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1068 Croxtall & Keam

Table III. Efficacy of oral raltegravir (RAL) 400 mg twice daily in adult patients (pts) with HIV type-1 (HIV-1) infection who have evidence of
viral replication and HIV-1 strains resistant to three classes of antiretroviral therapy (ART). Results from the pre-planned 16- and 48-week
analyses of all evaluable pts in the BENCHMRK trials. BENCHMRK-1 and BENCHMRK-2 are identically designed, ongoing, randomized,
double-blind, placebo (PL)-controlled, multicentre, phase III trials in pts with ART-resistant HIV-1 receiving RAL 400 mg twice daily plus
optimized background therapy (OBT) or PL twice daily plus OBT[26]
Study Treatment No. of Pts (%) with plasma HIV-1 RNA levelsa Mean change from baseline inb
regimen pts c
<400 <50 CD4+ cell counts plasma viral load
(copies/mL) (copies/mL)d (cells/mm3)d,e (log10 copies/mL)f
Results at 16 weeks
BENCHMRK 1 RAL + OBT 227 78*g 62* 83h -1.8h
PL + OBT 117 41 g
33 31 h
-0.8h
*g *
BENCHMRK 2 RAL + OBT 226 78 63 86 h
-1.9h
PL + OBT 118 43 g
36 40 h
-1.1h
Results at 48 weeks
BENCHMRK 1 RAL + OBT 227–231 74* 65* 120* -1.7*
PL + OBT 114–118 36 31 49 -0.8
BENCHMRK 2 RAL + OBT 216–228 71* 60* 98* -1.7*
PL + OBT 114–119 38 35 40 -0.8
BENCHMRK 1 and 2 RAL + OBT 439–459 72* 62* 109* -1.7*
PL + OBT 228–237 37 33 45 -0.8
a Treatment-related discontinuations at 16 weeks and pt noncompletion at 48 weeks were counted as virological failures. Pts were deemed
to have virological failure if HIV-1 RNA levels at or after week 16 were: (i) not reduced to <400 copies/mL or by >1 log10/mL from baseline;
(ii) increased by >1 log10/mL from the lower of two consecutive measurements; or (iii) ‡400 copies/mL on two consecutive measurements
following a prior level of <400 copies/mL.
b Observed lack of efficacy at 16 and 48 weeks counted as virological failure; data for treatment-related discontinuations and pt
noncompletion not reported.
c A total of 462 pts were randomized to RAL plus OBT and 237 to PL plus OBT in BENCHMRK 1 and 2. Where evaluable pt numbers varied
according to the statistical approach used for analysis, values are presented as a range.
d Secondary endpoint.
e Baseline values ranged from 102 to 140 cell/mm3.
f Baseline values ranged from 4.6 to 4.8 log10 copies/mL.
g Primary endpoint.
h Value estimated from a graph.
*
p < 0.001.

baseline in HIV-1 plasma viral load was -1.7 with patient non-completion counted as viro-
log10 copies/mL for raltegravir plus OBT versus logical failure; CD4+ cell counts increased by
-0.8 log10 copies/mL for placebo plus OBT re- 123 cells/mm3 versus 49 cells/mm3 (statistical
cipients, for both studies individually and the significance not reported).[43]
combined analysis, at week 48 (table III).[26] Prespecified subgroup analyses for potential
Preliminary data indicate that the anti- prognostic factors (using the observed lack of effi-
retroviral efficacy of raltegravir was maintained cacy as virological failure approach) suggested that
at 96 weeks in both BENCHMRK trials (avail- the efficacy of raltegravir plus OBT was not affected
able as an abstract).[43] Pooled data from both by baseline viral load (including HIV-1 RNA levels
studies showed that after 96 weeks, HIV-1 RNA >100 000 copies/mL) and CD4+ cell counts (inclu-
levels were reduced to <50 copies/mL in 58% of ding counts of <50 cells/mm3), various demographic
recipients of raltegravir plus OBT (n = 462) versus variables and viral subtypes.[14] However, a
33% of recipients of placebo plus OBT (n = 237), greater efficacy of raltegravir plus OBT over

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1069

placebo plus OBT was observed in patients with 9 RAL + OBT


PL + OBT
genotypic and phenotypic scores of 0–2. Posthoc 8
analysis indicated that the inclusion or non- 7
inclusion of darunavir and/or enfuvirtide as part 6

Patients (%)
of OBT had no effect on the improvement in 5
efficacy of raltegravir plus OBT over placebo plus 4
OBT, although there was an additional benefit 3
from the inclusion of these agents in both treat- 2
ment arms.[14] 1
0

es

ed n
se
nt
l

T
e ol

nc phi T
ro
as

i
ub
AL

AS
ou
id

r
5. Tolerability

la
ce Ele ste
e

n
er

y
ea st

El bilir
lc
ki

m
d
e
le
yc

ne ate

a
ol
o

at
cl

tin

l
ch

ta
ic
ed ch

ro

ev
tri

at

to
ut
L
The tolerability profile of raltegravir has been

l
ed

re
a

LD
cr

ed
o t
at

at
ed
reviewed previously.[9] This section provides an
ev

pa
ed

ev
at
El

at
at

El
ev

ed
u
ev
ev

ed
El
overview of the tolerability of raltegravir in com-

at
El
El

ev
El
bination with OBT in treatment-experienced
patients with HIV-1 infection in the ongoing Fig. 3. Tolerability profile of raltegravir (RAL). Combined analysis
of the most frequent (incidence of >3% in either treatment arm)
BENCHMRK trials described in section 4,[26] with laboratory abnormalities of grade 3 or 4 up to week 48 in the
additional supporting information provided from BENCHMRK (Blocking integrase in treatment Experienced patients
the manufacturer’s prescribing information.[18,19] with a Novel Compound against HIV: MeRcK) trials.[26] See table III
for trial design details. Patients were treated with raltegravir (RAL)
In general, raltegravir was well tolerated, with 400 mg twice daily plus optimized background therapy (OBT)
the majority of adverse events being of mild to [n = 462] or placebo (PL) twice daily plus OBT (n = 237). LDL = low-
density lipoprotein.
moderate severity.[26] Moreover, adverse event-
related discontinuations were uncommon, with
2% of patients receiving raltegravir plus OBT The most commonly reported adverse events
discontinuing treatment for this reason versus 3% regardless of causality for recipients of raltegravir
of recipients of placebo plus OBT.[18,19] 400 mg twice daily plus OBT (n = 507) versus
placebo twice daily plus OBT (n = 282) were
diarrhoea (17.6% vs 20.6%), nausea (11.2% vs
5 RAL + OBT 15.2%), headache (10.1% vs 12.4%) and pyrexia
PL + OBT (6.3% vs 11%) based on pooled safety data from
4
three randomized clinical trials.[19]
Patients (%)

3 Similarly, data from the combined BENCHMRK


2
studies showed that the most common drug-
related clinical adverse events of moderate to
1 severe intensity at 48 weeks were diarrhoea,
0 headache, nausea and fatigue, which were not
significantly different between the raltegravir
n

in
a

he

ea

ue
tio
oe

pa
c

ig
ac

plus OBT and placebo plus OBT treatment


da

au
rrh

ite
Fa
re

ea

N
ia

s
D

n-

groups (figure 2).[26] Injection-site reactions were


ite

io
s
n-

ct
je
io

considered to be related to the enfuvirtide com-


In
ct
je

ponent of OBT.[26]
In

Fig. 2. Tolerability profile of raltegravir (RAL). Combined analysis of Triglyceride elevation was the most common
common treatment-related adverse events of moderate to severe treatment-related laboratory abnormality in ral-
intensity up to week 48 in the BENCHMRK (Blocking integrase in
treatment Experienced patients with a Novel Compound against HIV: tegravir plus OBT recipients, followed by in-
MeRcK) trials.[26] See table III for trial design details. Patients were creased total cholesterol, creatine kinase, LDL
treated with RAL 400 mg twice daily plus optimized background
therapy (OBT) [n = 462] or placebo (PL) twice daily plus OBT
cholesterol and ALT levels, reduced neutrophil
(n = 237). count, and increased pancreatic amylase, total

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1070 Croxtall & Keam

bilirubin and AST levels which were not signi- Postmarketing surveillance has indicated a
ficantly different from the placebo plus OBT risk of depression and suicidal behaviours in pa-
treatment arm (figure 3).[26] Elevated creatine tients with pre-existing psychiatric illness. How-
kinase levels was the third most common labo- ever, the frequency of these events in relation to
ratory abnormality in raltegravir plus OBT re- raltegravir exposure is not clear.[18]
cipients and, although instances of myopathy and
rhabdomyolysis have been reported, the rela-
tionship of raltegravir treatment to these events 6. Dosage and Administration
is not known.[18,19]
Raltegravir is indicated in combination with
Following 48 weeks’ treatment in the com-
other ART agents for the treatment of HIV-1
bined BENCHMRK trials, 16 of the 462 ralte-
infection in treatment-experienced adult patients
gravir plus OBT recipients (3.5%) and 4 of the
who have evidence of viral replication and HIV-1
237 placebo plus OBT recipients (1.7%) had a
strains resistant to multiple ART agents.[18,19]
diagnosis of new, recurrent or progressive can-
The recommended dosage of raltegravir is 400 mg
cer.[26] The relative risk of cancer in the ralte-
twice daily administered orally with or without
gravir treatment arms was 1.54 (95% CI 0.50,
food.[18,19] Caution should be used when coad-
6.34) compared with the placebo arms.[26]
ministering raltegravir with strong inducers of
The types of cancer (Kaposi’s sarcoma, non-
UGT1A1 (e.g. rifampicin), as reduced plasma
Hodgkin’s lymphoma, anal squamous-cell carci-
concentrations of raltegravir may ensue (section
noma, rectal adenocarcinoma, hepatocellular
3.4). In the EU, a doubling of the dose of ralte-
carcinoma, skin cancer and laryngeal squamous-
gravir may be required if coadministration with
cell carcinoma) are those expected in patients
rifampicin cannot be avoided[19] and in the US,
with severe immunodeficiency, papillomavirus
the recommended raltegravir dosage is 800 mg
or active hepatitis B infection, and ongoing sur-
twice daily.[18] In the EU, coadministration of
veillance may be advisable. However, a recent
raltegravir with agents that increase gastric pH is
review of 1039 recipients of raltegravir plus
not recommended unless this is unavoidable.[19]
OBT pooled from several trials suggests that
Raltegravir should be used during pregnancy
malignancy rates associated with raltegravir
only if the potential maternal benefits outweigh
treatment are not significantly different from
fetal risk and is not recommended for breast-
that of comparator ART agents (available as an
feeding mothers.[18,19] Caution is advised when
abstract).[48]
administering raltegravir to patients with an in-
The tolerability profile of raltegravir plus OBT
creased risk of myopathy or rhabdomyolysis be-
was similar in treatment-experienced patients
cause of elevated creatine kinase levels, although
with HIV-1 infection and chronic active hepatitis
no direct link has been shown. Local prescribing
B and/or hepatitis C co-infections in the
information should be consulted for other con-
BENCHMRK trials.[18,19] However, worsening
traindications, warnings or recommendations.
from baseline of AST, ALT or total bilirubin
levels was 2- to 4-fold higher in raltegravir plus
OBT recipients with hepatitis co-infections than 7. Place of Raltegravir in the
in those without co-infections.[18,19] Management of HIV Infection in
When commencing combination ART, pa- Treatment-Experienced Patients
tients with severe immunodeficiency may develop
an inflammatory response to opportunistic in- The overall aim of the management of patients
fections (such as Mycobacterium avium complex, with HIV-1 infection is maximal suppression of
cytomegalovirus, pneumonia, tuberculosis or re- HIV-1 replication, preservation of immunological
activation of varicella zoster virus) when re- function and prevention of vertical transmis-
sponding to the initial phase of treatment, and sion.[5,49-51] Moreover, in patients with prior
these may require further intervention.[18] treatment experience exhibiting ART resistance,

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1071

the prevention of further selection of resistance Raltegravir is the first drug to be approved in
mutations is highly desirable.[49] the new class of ART integrase inhibitors. It is
Currently, first-line treatment regimens re- indicated in combination with other ART agents
commended for HIV-1-infected patients consist for the treatment of HIV-1 infection in treatment-
of three or more drugs from four classes of experienced adult patients who have evidence of
ART agents, which include NRTIs, NNRTIs, viral replication and HIV-1 strains resistant to
PIs and fusion inhibitors.[49-52] The implemen- multiple ART agents (section 6). One other in-
tation of these combination regimens has, in tegrase inhibitor, elvite- gravir, has been filed for
large part, been responsible for stabilizing the regulatory approval, and additional integrase
morbidity and mortality associated with HIV-1 inhibitors are in development.[8]
infections.[53] As the first integrase inhibitor, raltegravir
presents several advantages over other ART
However, these ART regimens are inherently
agents for treatment-experienced patients with
complex and have considerable adverse effects,
multiple-drug resistant HIV-1 infection. The
which presents difficulties for patient adherence
novel mechanism of action of raltegravir (section
resulting in suboptimal drug exposure and,
2.1) means that the potential for cross-resistance
thereby, an increased risk of the development of
with ART agents from other classes is much re-
viral resistance.[5] Drug-susceptibility profiles not
duced. Indeed, pharmacodynamic studies show
only play a crucial role in the selection of ART
that antiviral activity is maintained in human
regimen employed,[52] but also have a direct
cells in culture infected with a broad range of
bearing on efficacy outcomes in both ART-naive
HIV-1 clinical isolates, including primary isolates
and ART-experienced patients.[54] Indeed, the
from a variety of subtypes and isolates resistant
British HIV Association specifically recommends
to other ART agents (section 2.2). Moreover,
individualized ART regimens ‘‘in order to achieve
raltegravir inhibits integrase activity at nano-
maximum potency, durability, adherence, and
molar concentrations in vitro, and this activity
tolerability and to avoid long-term toxicities and
is enhanced additively or synergistically when
any likely drug interactions’’.[51]
ART agents from other classes are used in
For treatment-experienced patients with combination.
multiple-drug virological failure, the selection of Well designed, ongoing, phase III clinical
ART combination regimens that combine two or trials in adults (BENCHMRK-1 and -2) showed
three fully active drugs has, up until now, been ralte- gravir to be effective in the management of
compromised by the number of drug classes treatment-experienced patients with HIV-1 infec-
available and the degree of intraclass cross- tion resistant to multiple ART agents (section 4).
resistance that exists among these agents.[54] When added to an OBT regimen, raltegravir
The availability of novel ART agents from 400 mg twice daily reduced HIV-1 RNA levels to
alternative drug classes presents new oppor- <400 copies/mL and <50 copies/mL in a signi-
tunities for managing patients with HIV-1 ficantly greater number of patients than placebo
infection, particularly those with evidence of plus OBT after 48 weeks of treatment. Further-
multiple-drug resistance.[55] Furthermore, these more, mean CD4+ cell counts were increased by a
newer ART agents may provide an impetus significantly greater amount from baseline in re-
for redefining treatment guidelines for the man- cipients of raltegravir plus OBT compared with
agement of HIV-1 infections.[55] Indeed, the patients receiving placebo plus OBT. These trials
International AIDS Society USA panel now are scheduled to continue for at least 156 weeks.
recommends the addition of either an integrase As many as two-thirds of patients with HIV-1
inhibitor or a chemokine (C-C motif) receptor 5 infection have CD4+ cell counts of <200 cells/mm3
antagonist to combination therapy for HIV-1- at baseline,[51] which, typically, is predictive of a
infected patients with first-line failure of NNRTI- much poorer long-term prognosis.[5,49-51] How-
based regimens and multidrug resistance.[50] ever, subgroup analysis of the BENCHMRK

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1072 Croxtall & Keam

trials indicated that the greater efficacy of ralte- agent, patients’ susceptibility to resistance may be
gravir over placebo as part of an OBT regimen less likely than previously thought.[54]
was maintained not only in patients with lower More importantly, prespecified subgroup
CD4+ cell counts at baseline, but also in patients analyses of patients from the combined
with higher HIV-1 RNA levels and those receiv- BENCHMRK trials showed that those with
ing OBT regimens with genotypic or phenotypic higher HIV-1 RNA levels and less active OBT
sensitivity scores of 0–2 (section 4). Although regimens at baseline were at the greatest risk of
more active OBT regimens (genotypic or pheno- integrase mutations arising.[14] In other words,
typic sensitivity scores >0) were associated with profiling patient characteristics at baseline not
higher absolute response rates than those with only has a direct bearing on the potential inci-
a sensitivity score of 0, there appeared to be dence of resistance to raltegravir, but also re-
little advantage in adding raltegravir to OBT inforces the desirability of individualized ART
in patients with genotypic or phenotypic scores regimens for HIV-1-infected patients in order to
of ‡3.[14] optimize efficacy outcomes.[51]
A problem for all ART regimens is the develop- Currently, raltegravir is not approved for use
ment of drug resistance, leading to viral rebound in treatment-naive patients with HIV-1 infection.
and treatment failure. Moreover, continuing However, preliminary evidence from a phase II
ART in patients with drug failure is associated trial has suggested raltegravir may be as effica-
with the selection of additional resistance muta- cious and better tolerated than efavirenz when
tions.[54] This is particularly true for ART agents used in a combination regimen with tenofovir
with low genetic barriers to resistance, such as plus lamivudine in treatment-naive patients.[41,42]
the NNRTIs, lamivudine, emtricitabine and These observations are consistent with efficacy
enfuvirtide.[54] Expert opinion on the suscepti- outcomes and the tolerability profile for ralte-
bility of raltegravir recipients to developing re- gravir seen in treatment-experienced patients
sistance mutations is divided,[8,54,56] with some (section 4).
authors suggesting that there is a low genetic Preliminary data suggest that raltegravir
barrier to resistance[54] and some, using in vitro shows inhibitory activity in human cells in culture
serial passage models, suggesting that this barrier infected with a range of clinical isolates of HIV-2
is high.[8] (section 2.2). This viral strain is relatively in-
Resistance to raltegravir is usually associated sensitive to inhibition by conventional ART
with two or more key mutations within the inte- agents, and while inhibition by raltegravir is of
grase gene resulting in the amino acid substitu- interest, this observation needs to be supported
tions Q148H/K/R, N155H or Y143C/H/R.[54] with clinical data from patients with HIV-2 in-
The mutations Q148H and N155H are known fection before any meaningful conclusions may
specifically to decrease the susceptibility of be made.
HIV-1 to raltegravir in vitro by 7- to 14-fold. In Raltegravir was generally well tolerated in
the BENCHMRK trials, 105 of 462 raltegravir combination with other ART agents, with an
recipients exhibited virological failure after adverse event profile similar to that seen with
48 weeks of treatment; genotyping of 94 of these placebo plus OBT (section 5). The majority of
showed that 64 had a mutation within the inte- adverse events were mild to moderate in severity
grase gene known to confer resistance and 30 had and treatment discontinuations were uncommon.
genotypic changes of unknown significance. The most common systemic drug-related adverse
However, of the 64 patients with virological fail- events reported were diarrhoea, headache, nau-
ure and mutations known to confer resistance, 38 sea and fatigue. Furthermore, unlike other ART
had a genotypic sensitivity score of 0, and 20 had agents, raltegravir did not appear to be asso-
a genotypic score of 1.[14] Therefore, when ralte- ciated with clinically relevant hyperlipidaemia.
gravir is used within the recommended context of A numerically higher rate of new, recurrent or
combination with at least one other active ART progressive malignancies was observed in the

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1073

raltegravir plus OBT arms of the BENCHMRK combination of raltegravir and OBT will be a
trials versus the placebo plus OBT arms. How- valuable treatment option for this difficult-to-
ever, given the increased risk for malignancies treat patient group.
developing in this severely immunocompromised
patient population and other risk factors for
cancer, it is not known if the cancer diagnoses Disclosure
were related to raltegravir treatment.
The preparation of this review was not supported by any
Importantly, pharmacokinetic studies indicate
external funding. During the peer review process, the manu-
that raltegravir does not interact with the CYP facturer of the agent under review was offered an opportunity
system (section 3.3). As a consequence, drug inter- to comment on this article. Changes resulting from comments
actions with other ART agents that are metabo- received were made on the basis of scientific and editorial
merit.
lized by CYP isoenzymes are minimal (table II).
This is in contrast to elvitegravir, which is meta-
bolized by CYP3A4.[8] Like atazanavir, raltegravir
is metabolized via UGT-mediated glucuronida- References
1. Joint United Nations Programme on HIV/AIDS. 2008 report
tion, and although exposure to raltegravir is on the global AIDS epidemic [online]. Available from URL:
increased modestly when these agents are co- http://www.unaids.org/en/KnowledgeCentre/HIVData/
GlobalReport/2008/2008_Global_report.asp [Accessed 2008
administered, dosage adjustments are not re- Nov 26]
quired. Similarly, patients polymorphic for 2. WHO, UNAIDS, UNICEF. Towards universal access:
UGT1A1 do not require dosage adjustments of scaling up priority HIV/AIDS interventions in the health
sector. Progress report 2008 [online]. Available from URL:
raltegravir. http://www.who.int/hiv/pub/towards_universal_access_
Pharmacoeconomic analyses of raltegravir in report_2008.pdf [Accessed 2008 Nov 26]
treatment-experienced patients with HIV-1 in- 3. WHO. Global HIV prevalence has levelled off: improvements
in surveillance increase understanding of the epidemic,
fection are limited to a cost-effectiveness study resulting in substantial revisions to estimates [online]. Avail-
from the Swiss payer perspective published as an able from URL: http://www.who.int/mediacentre/news/
abstract.[57] The incremental cost-effectiveness releases/2007/pr61/en/print.html [Accessed 2008 Nov 26]
4. Shet A, Berry L, Mohri H, et al. Tracking the prevalence of
ratio of raltegravir plus OBT versus OBT alone transmitted antiretroviral drug-resistant HIV-1. J Aquir
was $US45 077 per quality-adjusted life year, Immune Defic Syndr 2006; 41 (4): 439-46
based on a raltegravir treatment period of 5 years 5. Dybul M, Fauci AS, Bartlett JG, et al. Guidelines for using
antiretroviral agents among HIV-infected adults and ado-
(2007 values).[57] Further pharmacoeconomic lescents: recommendations of the Panel on Clinical Prac-
analyses are warranted to clarify the cost effec- tices for Treatment of HIV. MMWR Recomm Rep 2002
tiveness of raltegravir-containing treatment regi- May 17; 51 (RR-7): 1-55
mens relative to other agents used in combination 6. Richman D, Morton S, Wrin T, et al. The prevalence of
antiretroviral drug resistance in the United States. AIDS
with OBT in these patients, and in other geo- 2004; 18 (10): 1393-401
graphical regions 7. Pommier Y, Johnson AA, Marchand C. Integrase inhi-
In conclusion, in clinical trials in treatment- bitors to treat HIV/AIDS. Nat Rev Drug Discov 2005; 4 (3):
236-48
experienced patients with HIV-1 infection and 8. Palmisano L. Role of integrase inhibitors in the treatment
evidence of viral replication, the addition of oral of HIV disease: expert review of anti-infective therapy.
raltegravir to an OBT regimen improved viro- Exp Rev Anti-Infect Ther 2007; 5 (1): 67-75
9. Croxtall JD, Lyseng-Williamson KA, Perry CM. Ralte-
logical and immunological responses at 16 and 48 gravir. Drugs 2008; 68 (1): 131-8
weeks to a greater extent than placebo plus OBT. 10. Summa V, Petrocchi A, Bonelli F, et al. Discovery of ralte-
Raltegravir therapy was generally well tolerated, gravir, a potent, selective orally bioavailable HIV-integrase
inhibitor for the treatment of HIV-AIDS infection. J Med
with a similar incidence of mild to moderate ad- Chem 2008; 51 (18): 5843-55
verse events in the treatment and placebo arms. 11. Marinello J, Marchand C, Mott BT, et al. Comparison of
The introduction of integrase inhibitors extends raltegravir and elvitegravir on HIV-1 integrase catalytic
the options available for managing treatment- reactions and on a series of drug-resistant integrase
mutants. Biochemistry (Mosc) 2008; 47 (36): 9345-54
experienced patients with multiple-drug-resistant 12. Danovich R, Ke Y, Wan H, et al. Raltegravir has similar
HIV-1 infection. Results to date suggest that the in vitro antiviral potency, clinical efficacy, and resistance

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
1074 Croxtall & Keam

patterns in B subtype and non-B subtype HIV-1 [abstract no. 30. Iwamoto M, Wenning LA, Petry AS, et al. Safety, toler-
TUAA0302]. 17th International AIDS Conference; 2008 Aug ability, and pharmacokinetics of raltegravir after single and
3-8; Mexico City multiple doses in healthy subjects. Clin Pharmacol Ther
13. Roquebert B, Damond F, Collin G, et al. HIV-2 integrase gene 2008; 83 (2): 293-9
polymorphism and phenotypic susceptibility of HIV-2 clin- 31. Kassahun K, McIntosh I, Cui D, et al. Metabolism and
ical isolates to the integrase inhibitors raltegravir and elvite- disposition in humans of raltegravir (MK-0518), an anti-
gravir in vitro. J Antimicrob Chemother 2008; 62 (5): 914-20 AIDS drug targeting the HIV-1 integrase enzyme. Drug
14. Cooper DA, Steigbigel RT, Gatell JM, et al. Subgroup and Metab Dispos 2007; 35 (9): 1657-63
resistance analyses of raltegravir for resistant HIV-1 in- 32. Iwamoto M, Wenning LA, Mistry GC, et al. Atazanavir
fection. N Engl J Med 2008; 359 (4): 355-65 modestly increases plasma levels of raltegravir in healthy
subjects. Clin Infect Dis 2008; 47 (1): 137-40
15. Charpentier C, Karmochkine M, Laureillard D, et al. Drug
resistance profiles for the HIV integrase gene in patients fail- 33. Iwamoto M, Wenning LA, Petry AS, et al. Minimal effects of
ing raltegravir salvage therapy. HIV Med 2008; 9 (9): 765-70 ritonavir and efavirenz on the pharmacokinetics of ralte-
gravir. Antimicrob Agents Chemother 2008; 52 (12): 4338-43
16. Malet I, Delelis O, Valantin MA, et al. Mutations associated
with failure of raltegravir treatment affect integrase sensi- 34. Iwamoto M, Kassahun K, Troyer MD, et al. Lack of a
tivity to the inhibitor in vitro. Antimicrob Agents Chemo- pharmacokinetic effect of raltegravir on midazolam:
ther 2008; 52 (4): 1351-8 in vitro/in vivo correlation. J Clin Pharmacol 2008; 48 (2):
209-14
17. Van Baelen K, Van Eygen V, Rondelez E, et al. Clade-
specific HIV-1 integrase polymorphisms do not reduce 35. Iwamoto M, Wenning, LA, Nguyen BY et al. Effects of
omeprazole on plasma levels of raltegravir. Clin Infect Dis.
raltegravir and elvitegravir phenotypic susceptibility.
Epub 2009 Jan 1
AIDS 2008; 22 (14): 1877-80
36. Markowitz M, Morales-Ramirez JO, Nguyen BY, et al. Anti-
18. Isentress (raltegravir): US prescribing information.
retroviral activity, pharmacokinetics, and tolerability of
Whitehouse Station (NJ): Merck and Co., Inc., 2009 MK-0518, a novel inhibitor of HIV-1 integrase, dosed as
19. European Medicines Agency. Raltegravir: summary of pro- monotherapy for 10 days in treatment-naive HIV-1-infected
duct characteristics [online]. Available from URL: http:// individuals. J Acquir Immune Defic Syndr 2006; 43 (5):
www.emea.europa.eu/humandocs/PDFs/EPAR/isentress/ 509-15
H-860-PI-en.pdf [Accessed 2009 May 20] 37. Iwamoto M, Hanley WD, Petry AS. Lack of a clinically
20. Rowley M. The discovery of raltegravir, an integrase in- important effect of moderate hepatic insufficiency and
hibitor for the treatment of HIV infection. Prog Med Chem severe renal insufficiency on raltegravir pharmacokinetics.
2008; 46: 1-28 Antimicrob Agents Chemother 2009 May; 53 (5): 1747-52
21. Evering TH, Markowitz M. Raltegravir (MK-0518): an in- 38. Wenning L, Hanley W, Brainard D, et al. Effect of rifampin,
tegrase inhibitor for the treatment of HIV-1. Drugs Today a potent inducer of drug metabolizing enzymes, on the
2007; 43 (12): 865-77 pharmacokinetics of raltegravir. Antimicrob Agents Che-
22. Hazuda DJ, Felock P, Witmer M, et al. Inhibitors of strand mother. Epub 2009 May 11
transfer that prevent integration and inhibit HIV-1 re- 39. Hanley W, Wenning L, Moreau A, et al. Effect of tiprana-
plication in cells. Science 2000; 287 (5453): 646-50 vir + ritonavir on pharmacokinetics of raltegravir. Anti-
23. Roquebert B, Blum L, Collin G, et al. Selection of the Q148R microb Agents Chemother. Epub 2009 Apr 27
integrase inhibitor resistance mutation in a failing raltegravir 40. McKeage K, Perry C, Keam S. Darunavir: a review of its use
containing regimen [letter]. AIDS 2008; 22 (15): 2045-6 in the management of HIV infection in adults. Drugs 2009;
69 (4): 477-503
24. Hazuda DJ, Miller MD, Nguyen BY. Resistance to the HIV-
integrase inhibitor raltegravir: analysis of protocol 005, a 41. Markowitz M, Nguyen B-Y, Gotuzzo E, et al. Rapid and
phase II study in patients with triple-class resistant HIV-1 durable antiretroviral effect of the HIV-1 integrase
infection [abstract no. 8]. Antiviral Therapy 2007 June inhibitor raltegravir as part combination therapy in
12-16; 12 (5): S10 treatment-naive patients with HIV-1 infection. J Acquir
Immune Defic Syndr 2007; 46 (2): 125-33
25. Garrett N, Xu L, Smit E, et al. Raltegravir treatment re-
sponse in an HIV-2 infected patient: a case report [letter]. 42. Markowitz M, Nguyen BY, Gotuzzo E, et al. Sustained
AIDS 2008 May 31; 22 (9): 1091-2 antiretroviral efficacy of raltegravir as part of combination
ART in treatment-naive HIV-1 infected patients: 96-week
26. Steigbigel RT, Cooper DA, Kumar PN, et al. Raltegravir data [abstract no. TUAB0102]. 17th International AIDS
with optimized background therapy for resistant HIV-1 Conference; 2008 Aug 3-8; Mexico City
infection. N Engl J Med 2008; 359 (4): 339-54
43. Steigbigel R, Cooper D, Eron J, et al. 96-week results from
27. Iwamoto M, Kost JT, Mistry GC, et al. Raltegravir tho- BENCHMRK 1 and 2, phase III studies of raltegravir in
rough QT/QTc study: a single supratherapeutic dose of patients failing ART with triple-class-resistant HIV [ab-
raltegravir does not prolong the QTcF interval. J Clin stract no. K-105]. 16th Conference on Retroviruses and
Pharmacol 2008; 48 (6): 726-33 Opportunistic Infections; 2009 Feb 9-11; Montreal (QC)
28. Wenning LA, Petry A, Kost JT, et al. Pharmacokinetics of 44. Grinsztejn B, Nguyen BY, Katlama C, et al. Safety and
raltegravir in individuals with UGT1A1polymorphisms. efficacy of the HIV-1 integrase inhibitor raltegravir
Clin Pharmacol Ther. Epub 2009 Mar 11 (MK-0518) in treatment-experienced patients with multi-
29. Wenning L, Anderson M, Petry A, et al. Raltegravir (RAL) drug-resistant virus: a phase II randomised controlled trial.
dose proportionality and effect of food [abstract no. Lancet 2007; 369 (9569): 1261-9
H-1046]. 47th Interscience Conference on Antimicrobial 45. Yazdanpanah Y, Fagard C, Descamps D, et al. High rate of
Agents and Chemotherapy; 2007 Sep 17-20; Chicago (IL) virologic success with raltegravir plus etravirine and

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)
Raltegravir: A Review 1075

darunavir/ritonavir in treatment-experienced patients with 52. European AIDS Clinical Society (EACS). Guidelines for the
multidrug-resistant virus: results of the ANRS 139 TRIO clinical management and treatment of HIV infected adults
trial [abstract no. THAB0406]. 17th International AIDS in Europe [online]. Available from URL: http://www.
Conference; 2008 Aug 3-8; Mexico City europeanaidsclinicalsociety.org/guidelinespdf/1_Treatment_
46. Harris M, Larsen G, Montaner JS. Outcomes of multidrug- of_HIV_Infected_Adults.pdf [Accessed 2009 Mar 26]
resistant patients switched from enfuvirtide to raltegravir 53. Palella FJ, Delaney K, Moorman A, et al. Declining mor-
within a virologically suppressive regimen. AIDS 2008 Jun bidity and mortality among patients with advanced human
19; 22 (10): 1224-6 immunodeficiency virus infection. N Engl J Med 1998; 338
47. De Castro N, Braun J, Charreau I, et al. Switch from en- (13): 853-60
fuvirtide (E) to raltegravir (R) in highly treatment-experi- 54. Hirsch MS, Gunthard HF, Schapiro JM, et al. Anti-
enced HIV-1 infected patients: a randomized open-label retroviral drug resistance testing in adult HIV-1 infection:
non-inferiority trial (Easier - ANRS 138) [abstract no. 572]. 2008 recommendations of an International AIDS Society-
16th Conference on Retroviuses and Opportunistic Infec- USA panel. Clin Infect Dis 2008; 47 (2): 266-85
tions; 2009 Feb 8-11; Montreal (QC) 55. Daar E. Emerging resistance profiles of newly approved
48. Cooper D, Steigbigel R, Lennox J, et al. Review of cancer antiretroviral drugs. Top HIV Med 2008; 16 (4): 110-6
incidence in raltegravir (RAL) clinical trials [abstract no. 56. Hughes A, Barber T, Nelson M. New treatment options for
R-106]. 16th Conference on Retroviruses and Opportu- HIV salvage patients: an overview of second generation
nistic Infections; 2009 Feb 8-11; Montreal (QC) PIs, NNRTIs, integrase inhibitors and CCR5 antagonists.
49. DHHS panel on antiretroviral guidelines for adults and ado- J Infect 2008; 57 (1): 1-10
lescents: a working group of the Office of AIDS Research 57. Elbasha E, Szucs T, Chaudhary M, et al. Cost-effectiveness
Advisory Council (OARAC). Guidelines for the use of anti- analysis of raltegravir in treatment-experienced HIV-1
retroviral agents in HIV-1-infected adults and adolescents infected patients in Switzerland [abstract no. TUPDD203].
[online]. Available from URL: http://aidsinfo.nih.gov/con 17th International AIDS Conference; 2008 Aug 3-8;
tentfiles/AdultandAdolescentGL.pdf [Accessed 2008 Nov 26] Mexico City
50. Hammer SM, Eron Jr JJ, Reiss P, et al. Antiretroviral
treatment of adult HIV infection: 2008 recommendations
of the International AIDS Society-USA panel. JAMA
2008; 300 (5): 555-70 Correspondence: Jamie D. Croxtall, Wolters Kluwer
51. Gazzard BG. British HIV Association guidelines for the Health | Adis, 41 Centorian Drive, Private Bag 65901,
treatment of HIV-1-infected adults with antiretroviral Mairangi Bay, North Shore 0754, Auckland, New Zealand.
therapy 2008. HIV Med 2008; 9 (8): 563-608 E-mail: demail@adis.co.nz

ª 2009 Adis Data Information BV. All rights reserved. Drugs 2009; 69 (8)

S-ar putea să vă placă și