ArticlePDF Available

Efficacy of the Protease Inhibitors Tipranavir plus Ritonavir in Treatment-Experienced Patients: 24-Week Analysis from the RESIST-1 Trial

Authors:

Abstract and Figures

Improved treatment options are needed for patients infected with multidrug-resistant human immunodeficiency virus type 1 (HIV-1). The nonpeptidic protease inhibitor tipranavir has demonstrated antiviral activity against many protease inhibitor-resistant HIV-1 isolates. The Randomized Evaluation of Strategic Intervention in multi-drug reSistant patients with Tipranavir (RESIST-1) trial is an ongoing, open-label study comparing the efficacy and safety of ritonavir-boosted tipranavir (TPV/r) with an investigator-selected ritonavir-boosted comparator protease inhibitor (CPI/r) in treatment-experienced, HIV-1-infected patients. Six hundred twenty antiretroviral-experienced patients were treated at 125 sites in North America and Australia. Before randomization, all patients underwent genotypic resistance testing, which investigators used to select a CPI/r and an optimized background regimen. Patients were randomized to receive TPV/r or CPI/r and were stratified on the basis of preselected protease inhibitor and enfuvirtide use. Treatment response was defined as a confirmed reduction in the HIV-1 load of > or = 1 log10 less than the baseline level without treatment change at week 24. Mean baseline HIV-1 loads and CD4+ cell counts were 4.74 log10 copies/mL and 164 cells/mm3, respectively. At week 24, a total of 41.5% of patients in the TPV/r arm and 22.3% in the CPI/r arm had a > or = 1-log10 reduction in the HIV-1 load (intent-to-treat population; P<.0001). Mean increases in the CD4+ cell count of 54 and 24 cells/mm3 occurred in the TPV/r and CPI/r groups, respectively. Adverse events were slightly more common in the TPV/r group and included diarrhea, nausea, and vomiting. Elevations in alanine and aspartate aminotransferase levels and in cholesterol/triglyceride levels were more frequent in the TPV/r group. TPV/r demonstrated superior antiviral activity, compared with investigator-selected, ritonavir-boosted protease inhibitors, at week 24 in treatment-experienced patients with multidrug-resistant HIV-1 infection.
Content may be subject to copyright.
HIV/AIDS CID 2006:43 (15 November) 1337
HIV/AIDSMAJOR ARTICLE
Efficacy of the Protease Inhibitors Tipranavir
plus Ritonavir in Treatment-Experienced Patients:
24-Week Analysis from the RESIST-1 Trial
Joseph Gathe,
1
David A. Cooper,
10
Charles Farthing,
2
Dushyantha Jayaweera,
3
Dorece Norris,
5
Gerald Pierone, Jr.,
6
Corklin R. Steinhart,
4
Benoit Trottier,
8
Sharon L. Walmsley,
9
Cassy Workman,
11
Geoffrey Mukwaya,
7
Veronika Kohlbrenner,
7
Catherine Dohnanyi,
7
Scott McCallister,
7
and Douglas Mayers,
7
for the RESIST-1 Study
Group
a
1
Therapeutic Concepts, Houston, Texas;
2
AIDS Healthcare Foundation, Los Angeles, California;
3
University of Miami School of Medicine and
4
Steinhart Medical Associates, Miami,
5
Comprehensive Research Institute, Tampa, and
6
AIDS Research and Treatment Center of the Treasure
Coast, Fort Pierce, Florida;
7
Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut;
8
Centre de Recherche en Toxicologie de
l’Environnement, Universite du Quebec a Montreal, Montreal, and
9
University of Toronto, Ontario, Canada;
10
National Centre in HIV Epidemiology
and Clinical Research, University of New South Wales, and
11
AIDS Research Initiative/Ground Zero Medical, Sydney, Australia
(See the article by Cahn et al. on pages 1347–56)
Background. Improved treatment options are needed for patients infected with multidrug-resistant human
immunodeficiency virus type 1 (HIV-1). The nonpeptidic protease inhibitor tipranavir has demonstrated antiviral
activity against many protease inhibitor–resistant HIV-1 isolates. The Randomized Evaluation of Strategic Inter-
vention in multi-drug reSistant patients with Tipranavir (RESIST-1) trial is an ongoing, open-label study comparing
the efficacy and safety of ritonavir-boosted tipranavir (TPV/r) with an investigator-selected ritonavir-boosted
comparator protease inhibitor (CPI/r) in treatment-experienced, HIV-1–infected patients.
Methods. Six hundred twenty antiretroviral-experienced patients were treated at 125 sites in North America
and Australia. Before randomization, all patients underwent genotypic resistance testing, which investigators used
to select a CPI/r and an optimized background regimen. Patients were randomized to receive TPV/r or CPI/r and
were stratified on the basis of preselected protease inhibitor and enfuvirtide use. Treatment response was defined
as a confirmed reduction in the HIV-1 load of 1log
10
less than the baseline level without treatment change at
week 24.
Results. Mean baseline HIV-1 loads and CD4
+
cell counts were 4.74 log
10
copies/mL and 164 cells/mm
3
,
respectively. At week 24, a total of 41.5% of patients in the TPV/r arm and 22.3% in the CPI/r arm had a 1-
log
10
reduction in the HIV-1 load (intent-to-treat population; ). Mean increases in the CD4
+
cell countP ! .0001
of 54 and 24 cells/mm
3
occurred in the TPV/r and CPI/r groups, respectively. Adverse events were slightly more
common in the TPV/r group and included diarrhea, nausea, and vomiting. Elevations in alanine and aspartate
aminotransferase levels and in cholesterol/triglyceride levels were more frequent in the TPV/r group.
Conclusions. TPV/r demonstrated superior antiviral activity, compared with investigator-selected, ritonavir-
boosted protease inhibitors, at week 24 in treatment-experienced patients with multidrug-resistant HIV-1 infection.
Protease inhibitor (PI)–based combination antiretro-
viral therapy has produced significant decreases in mor-
bidity and mortality in patients with HIV-1 infection
[1]; however, treatment failure still occurs [2–5] as a
Received 23 February 2006; accepted 6 July 2006; electronically published 17
October 2006.
a
Members of the study group are listed at the end of the text.
Reprints or correspondence: Dr. Joseph Gathe, Therapeutic Concepts, 4900
Fannin St., PA Houston, TX 77004 (drgathe@josephgathe.com).
Clinical Infectious Diseases 2006; 43:1337–46
2006 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2006/4310-0019$15.00
result of poor tolerability, lack of adherence, and chal-
lenging dosing regimens, all of which can lead to viral
resistance [6–8]. Such issues may limit options for fu-
ture therapy [9–11], particularly for patients who ex-
perience triple–drug class virologic failure and who may
be at increased risk of death (3-year mortality rate,
15%) [12]. Therefore, despite the undisputed benefits
of modern antiretroviral therapy regimens, newer
agents with activity against drug-resistant HIV-1 are
needed.
Tipranavir is a nonpeptidic PI of the dihydropyrone
sulfonamide class with a structure differing from that
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
1338 CID 2006:43 (15 November) HIV/AIDS
of peptidic PIs, and in vitro studies have shown that it may be
effective against virus strains that are resistant to available pep-
tidic PIs [13–18]. Tipranavir is rapidly and extensively metab-
olized through the cytochrome P450 isoenzyme 3A [19–22]
and must be coadministered with ritonavir to attain therapeutic
concentrations [23]. Ritonavir-boosted tipranavir (TPV/r) has
demonstrated potent antiviral activity in both treatment-naive
and treatment-experienced patients [24, 25].
The Randomized Evaluation of Strategic Intervention in
multi-drug reSistant patients with Tipranavir (RESIST-1) trial
is an ongoing, open-label, phase III multicenter study designed
to evaluate the efficacy and safety of TPV/r (500 mg/200 mg
twice per day, selected on the basis of phase II dose-ranging
studies) and an investigator-selected, ritonavir-boosted, stan-
dard-of-care comparator PI (CPI/r), when given with an op-
timized background regimen to treatment-experienced, HIV-
1–infected patients. A similar study (RESIST-2) has been
conducted in Europe and Latin America [26]. The results of
the 24-week interim analysis of RESIST-1 are presented here.
METHODS
Patients. HIV-1–infected adult patients were screened for
baseline HIV-1 RNA levels 1000 copies/mL (i.e., the mini-
mum level required for genotyping); documented baseline ge-
notypic resistance demonstrating 1 primary PI mutation (co-
dons 30N, 46I/L, 48V, 50V, 82A/F/L/T, 84V, or 90M) [27]; no
more than 2 PI resistance-associated mutations at codons 33,
82, 84, or 90; at least 3 consecutive months of experience with
all antiretroviral therapy agents (nucleoside reverse-transcrip-
tase inhibitors [NRTIs], nonnucleoside reverse-transcriptase in-
hibitor [NNRTIs], and PIs); experience with 2 PI-based reg-
imens, one of which had to be the regimen at baseline. There
was no restriction with regard to the CD4
+
cell count. Safety
screening laboratory values of the Division of AIDS of the
National Institutes of Health of grade 2 were exclusionary
(grade 2 was allowed for total cholesterol and triglyceride
levels). Other exclusion criteria included interruption in the
antiretroviral treatment regimen for 7 consecutive days within
3 months of screening, prior tipranavir use, a positive preg-
nancy test result, or breast-feeding. Patients were excluded if
they required other investigational medications, immunomo-
dulatory drugs, or ethinyl estradiol within 30 days of study
entry; if they were actively abusing substances that affected
protocol participation; if they had an unacceptable medical
history, as determined by the investigator (e.g., on the basis of
exclusionary chest radiograph or electrocardiograph findings);
or if they were unlikely to survive for 12 months. The protocol
and written informed consent forms were reviewed and ap-
proved by an institutional review board or ethics committee
before patients entered the study.
Study design. This ongoing, randomized, open-label, phase
III study is being conducted at 125 sites in the United States,
Canada, and Australia for a treatment period of 96 weeks. The
main efficacy end point is treatment response, defined as the
proportion of patients with a reduction in the HIV-1 load of
1log
10
after 24 weeks (confirmed by 2 consecutive measure-
ments), without having experienced virologic failure, discon-
tinued treatment with the study PI, introduced new antiret-
roviral agents because of a lack of treatment efficacy, neglected
to maintain follow-up, or died. Other end points included the
change from the baseline value in plasma HIV-1 RNA levels,
achievement of an HIV-1 load
!400 or !50 copies/mL, changes
in the CD4
+
cell count during treatment, and safety measures.
Treatment. Before randomization, investigators selected
both a CPI/r and an NRTI-based and NNRTI-based optimized
background regimen (manufacturer’s recommended dosages)
for each patient on the basis of genotypic resistance screening
findings and the patient’s antiretroviral medication history. An
expert resistance panel was available to help select the CPI/r.
Patients were randomized to receive TPV/r or a preselected
CPI/r (lopinavir-ritonavir, 400 mg/100 mg twice per day; in-
dinavir-ritonavir, 800 mg/100 mg twice per day; saquinavir-
ritonavir, 1000 mg/100 mg or 800 mg/200 mg twice per day;
or amprenavir-ritonavir, 600 mg/100 mg twice per day). Ran-
domization was stratified by both the preselected PI and the
use of enfuvirtide. Tipranavir (250-mg capsules) was supplied
by Boehringer Ingelheim, and CPIs and ritonavir were com-
mercially acquired.
Changes to the treatment regimen were permitted only for
reasons of toxicity and/or intolerance to the non-PI compo-
nents of the regimen. After week 8, patients in the CPI/r arm
had the option of discontinuing the assigned CPI because of a
lack of initial virologic response (defined as a decrease in the
HIV-1 load of
!0.5 log
10
from baseline or failure to achieve an
HIV-1 load of
!100,000 copies/mL despite having a 0.5-log
10
decrease) or confirmed virologic failure (defined as an HIV-1
load of
!1log
10
less than the baseline level confirmed on 2
consecutive assays or as 1 HIV-1 load of
!1log
10
less than the
baseline value followed by a permanent discontinuation of ther-
apy) to receive TPV/r as part of a separate rollover study. This
option was only permitted for participants who experienced
confirmed HIV-1 load failure who had a measurable CPI blood
concentration. Trough plasma drug concentrations for all PIs
and ritonavir were determined at weeks 2, 4, and 28 (i.e., visits
4, 5, and 8). The ratio of the geometric mean trough plasma
concentration for the PI to the IC
50
(both unadjusted and pro-
tein adjusted) of the HIV isolate at study entry was calculated
and correlated to subsequent virologic response.
Sample analysis. Plasma HIV-1 RNA levels were measured
using the Amplicor HIV-1 Monitor Assay, version 1.5 (Roche),
or the UltraSensitive method, version 1.5 (Roche). CD4
+
cell
counts were measured using standard flow cytometry. All tests
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
HIV/AIDS CID 2006:43 (15 November) 1339
were conducted by Covance Central Laboratory Services (In-
dianapolis, IN). HIV genotype resistance assessments were per-
formed using the TruGene method, version 1.0, at screening;
at weeks 4, 8, 16, and 24; and at end of treatment. Phenotypic
drug resistance was determined in a randomly selected subset
of 250 patients by Virco NV (Mechelen, Belgium) using Virco’s
Antivirogram assay. The Division of AIDS adverse events scale
was used to grade adverse event intensity and several laboratory
abnormalities, and the Common Toxicity Criteria scale was
used to grade cholesterol levels.
Statistical analysis. Differences in treatment response at
week 24 were analyzed by a 2-sided 95% CI, which was adjusted
for preselected PI and enfuvirtide use [28]. A sample size of
247 patients per treatment arm provided 90% power to detect
15% superiority of tipranavir over CPIs in virologic response
at 24 weeks, using a 2-sided Fisher’s exact test. All efficacy
analyses use the intent-to-treat, noncompleter-considered-fail-
ure analysis, in which missing values associated with premature
discontinuation of treatment were considered to indicate treat-
ment failure. Changes in the HIV-1 load and CD4
+
cell count
over time were evaluated using the last-observation-carried-
forward analysis.
RESULTS
Baseline Patient Characteristics
Between January 2003 and April 2004, a total of 1406 patients
were screened for the RESIST-1 trial, and 630 patients were
randomized (figure 1). Baseline characteristics were comparable
between groups (table 1). The median number of genotypically
available antiretrovirals in the optimized background regimen
was 2 in the TPV/r group and 1 in the CPI/r group, and more
patients in the TPV/r arm exhibited resistance to their prese-
lected PI (54.3% vs. 60.5%). A higher proportion of patients
in the CPI/r group than the TPV/r group had hepatitis C virus
coinfection (7.4% vs. 3.2%).
Baseline Resistance Characteristics
Baseline genotypes identified a mean of 2 primary protease gene
mutations from among 30N, 46I/L, 48V, 50V, 82A/F/L/T, 84V,
or 90M, as well as a mean of 15 total protease gene mutations
or polymorphisms. The median baseline phenotypic fold-
change in susceptibility to tipranavir for the random sample
of viral isolates was 1.9 times wild-type IC
50
, compared with
the following CPI IC
50
values for the same specimens: 77.8-
fold for lopinavir, 39.0-fold for indinavir, 27.2-fold for saqui-
navir, and 12.2-fold for amprenavir. The expert resistance panel
was consulted for optimized background regimen selection in
154 (25%) of 620 cases, with 114 (74%) of their 154 recom-
mendations being implemented. The optimized background
regimen included a median of 1 genotypically active antiret-
roviral, including enfuvirtide (range, 0–4 antiretrovirals). The
most common optimized background regimen (not including
the CPI), which was received by 158 patients (25.5%), was 2
NRTIs. On the basis of the genotype test results, 176 patients
(56.6%) in the TPV/r group and 183 patients (59.2%) in the
CPI/r group were assigned a new PI.
Patient Disposition
Overall, 263 patients (84.6%) and 151 patients (48.9%) in the
TPV/r and CPI/r groups, respectively, completed 24 weeks of
treatment (figure 1). More patients in the TPV/r arm (199
patients [64.0%]) than in the CPI/r arm (136 patients [44.0%])
received study medication for at least 24 weeks. This resulted
in a greater number of patient-exposure-years for the TPV/r
arm (133.3 years) than for the CPI/r arm (115.8 years).
Efficacy End Points
Treatment response. Patients who received TPV/r demon-
strated a significantly higher treatment response rate (according
to intent-to-treat, noncompleter-considered-failure analysis),
compared with CPI/r recipients (41.5% vs. 22.3%; )P
! .0001
(figure 2A). After response rates for the 2 arms were adjusted
for different CPIs and enfuvirtide use, TPV/r had an 18.4%
higher treatment response rate, compared with CPI/r (95% CI,
11.4%–25.3%). When treatment response rate was analyzed by
the individual CPI, TPV/r was superior to each CPI (lopinavir
group, 37.2% for tipranavir vs. 24.1% for lopinavir [
P p
]; saquinavir group, 45.3% for tipranavir vs. 18.8% for
.0069
saquinavir [ ]; amprenavir group, 52.4% for tipran-
P p .0005
avir vs. 24.4% for amprenavir [ ]; and indinavir
P p .0057
group, 50.0% for tipranavir vs. 7.7% for indinavir [ ]).
P p .02
A superior treatment response was also reported with the
TPV/r group in patients with 3–4 or 5–6 primary PI-associated
mutations at week 24, compared with the CPI/r group (3–4
mutations, 78 [42.9%] of 182 patients vs. 30 [15.5%] of 194
patients; 5–6 mutations, 2 [66.7%] of 3 patients vs. 1 [33.3%]
of 3 patients). Furthermore, patients with 1–2 PI mutations at
codons 33, 82, 84, or 90 in the TPV/r group had greater treat-
ment response values than did comparable patients in the CPI/
r group (1 mutation, 42 [46.2%] of 91 patients vs. 25 [32.5%]
of 77 patients; 2 mutations, 81 [42.2%] of 192 patients vs. 36
[17.6%] of 204 patients). At week 24, when the treatment re-
sponse was evaluated in 182 TPV/r recipients with an available
baseline phenotype, there was a 47% response rate (54 of 115
patients) in patients with a baseline IC
50
fold-change of !3,
compared with 28% (19 of 67 patients) in those with a baseline
IC
50
fold-change of 3–8. Patients with a baseline HIV-1 load
of
!10,000 copies/mL were more likely to achieve a treatment
response than were those with higher baseline HIV-1 loads
(table 2).
Other efficacy end points. The 24-week mean reduction in
HIV-1 load from baseline was significantly greater in the TPV/
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
1340 CID 2006:43 (15 November) HIV/AIDS
Figure 1. Patient disposition in a study of the efficacy of the protease inhibitors tipranavir and ritonavir. CPI/r, ritonavir-boosted comparator protease
inhibitor; TPV/r, ritonavir-boosted tipranavir.
r group (1.28 log
10
) than in the CPI/r group (0.64 log
10
;
) (figure 2B). More TPV/r recipients achieved an un-
P
! .001
detectable HIV-1 load (cutoff of
!400 copies/mL, 34.7%; cutoff
of
!50 copies/mL, 25.1%), compared with CPI/r recipients
(cutoff of
!400 copies/mL, 16.5%; cutoff of !50 copies/mL,
10.0%; ) (figure 2C). Furthermore, among TPV/r and
P
! .0001
CPI/r recipients who received enfuvirtide, the proportion of
patients with an undetectable HIV-1 load increased during
treatment (for TPV/r recipients, 47.1% and 32.8% for cutoff
values of
!400 and !50 copies/mL, respectively; for CPI/r re-
cipients, 21.9% and 14.3% for cutoff values of
!400 and !50
copies/mL, respectively). The mean increase from baseline in
the CD4
+
cell count was significantly greater in the TPV/r group
than in the CPI/r group (+54 vs. +24 cells/mm
3
; ) (fig-
P
! .001
ure 2D).
The treatment response rate for TPV/r increased from 11.6%
(5 of 43 patients), when the optimized background regimen
contained no genotypically susceptible drugs, to 57.6% (19 of
33 patients), when there were 3 genotypically susceptible
drugs. Response rates with CPI/r ranged from 13.2% (7 of 53
patients), when there were no genotypically active drugs, to
41.0% (16 of 39 patients), when there were
13 active drugs.
The addition of enfuvirtide to the optimized background reg-
imen improved treatment response, from 31.3% to 58.0% in
the TPV/r group and from 18.6% to 29.5% in the CPI/r group.
The effect of enfuvirtide on treatment response was enhanced
in TPV/r-treated patients who were enfuvirtide naive (66.7%),
compared with enfuvirtide-experienced patients (31.0%).
Trough plasma concentrations were analyzed at weeks 2 and
4. Detectable plasma PI concentrations had to be documented
before approval was granted for a switch of a patient from the
CPI/r group to the TPV/r group. There was no difference in
mean drug concentration measurements for patients who
changed treatment group versus those who did not.
Safety
All 620 treated patients were included in the safety analysis.
Most patients experienced at least 1 adverse event (90.7% of
TPV/r recipients and 86.4% of CPI/r recipients) (table 3). The
majority of adverse events in the 2 groups were graded as mild
(in 78.5% of TPV/r recipients and 75.7% of CPI/r recipients)
or moderate (in 59.8% of TPV/r recipients and 53.4% of CPI/
r recipients) in intensity. In the TPV/r group, adverse events
leading to discontinuation (2 patients) were nausea, diarrhea,
increased alanine aminotransferase (ALT) levels, vomiting, cer-
ebrovascular accident, fatigue, pyrexia, and sepsis, whereas in
the CPI/r group, adverse events leading to discontinuation (2
patients) were nausea, diarrhea, vomiting, and abdominal pain
(
!2% of patients in all cases).
Serious adverse events were experienced by 55 patients
(17.7%) in the TPV/r arm and 42 patients (13.6%) in the CPI/
r arm; fever (TPV/r group, 1.9%; CPI/r group, 1.3%), diarrhea
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
HIV/AIDS CID 2006:43 (15 November) 1341
Table 1. Baseline demographic characteristics of patients.
Characteristic
Treatment group
Overall
(n p 620)
TPV/r arm
(n p 311)
CPI/r arm
(n p 309)
Male sex 565 (91.1) 278 (89.4) 287 (92.9)
Age, median years (range) 44 (24 –80) 45 (24–80) 43 (28–70)
Median no. of NRTIs used (range) 6 (2–8) 6 (2–8) 6 (2–8)
Median no. of NNRTIs used (range) 2 (0–3) 2 (0–3) 1 (0–3)
Median no. of PIs used (range) 4 (1–7) 4 (1–7) 4 (1–7)
HIV-1 load, median log
10
copies/mL (range) 4.83 (2.01–6.31) 4.81 (2.34–6.13) 4.84 (2.01–6.31)
CD4
+
cell count, median cells/mm
3
(range) 123 (1–1184) 123 (1–860) 123 (1–1184)
Fusion inhibitor use 76 (12.3) 39 (12.5) 37 (12.0)
History of AIDS-defining illnesses 421 (67.9) 209 (67.2) 212 (68.6)
Median no. of primary protease mutations (range) 2.7 (0–5) 2.7 (0–5) 2.7 (0–5)
No. of protease mutations at 33, 82, 84, and 90
a
0 24 (3.9) 11 (3.5) 13 (4.2)
1 168 (27.1) 91 (29.3) 77 (24.9)
2 396 (63.9) 192 (61.7) 204 (66.0)
New PI selected that was not part of screening treatment regimen 359 (57.9) 176 (56.6) 183 (59.2)
Resistance to preselected PI
Susceptible
b
49 (7.9) 21 (6.8) 28 (9.1)
Possible resistance
c
214 (34.5) 120 (38.6) 94 (30.4)
Resistance
d
356 (57.4) 169 (54.3) 187 (60.5)
Median no. of genotypically available ARVs in the optimized back-
ground regimen (range)
e
1 (0–4) 2 (0–4) 1 (0–4)
Treatment assignment
Lopinavir 378 (61.0) 191 (61.4) 187 (60.5)
Indinavir 27 (4.4) 14 (4.5) 13 (4.2)
Saquinavir 128 (20.6) 64 (20.6) 64 (20.7)
Amprenavir 87 (14.0) 42 (13.5) 45 (14.6)
Enfuvirtide 224 (36.1) 119 (38.3) 105 (34.0)
Hepatitis virus coinfection status
HBsAg and HCV RNA negative 559 (90.2) 286 (92.0) 273 (88.3)
HBsAg positive and HCV RNA negative 26 (4.2) 14 (4.5) 12 (3.9)
HBsAg negative and HCV RNA positive 31 (5.0) 10 (3.2) 21 (6.8)
HBsAg and HCV RNA positive 2 (0.3) 0 2 (0.6)
Missing 2 (0.3) 1 (0.3) 1 (0.3)
Hepatitis B antibody positive 335 (54.0) 171 (55.0) 164 (53.1)
NOTE. Data are no. (%) of subjects, unless otherwise indicated. ARV, antiretroviral; CPI/r, ritonavir-boosted comparator protease inhibitor;
HBsAg, hepatitis B surface antigen; HCV, hepatitis C virus; NNRTI, nonnucleoside reverse-transcriptase inhibitor; NRTI: nucleoside reverse-
transcriptase inhibitor; TPV/r, ritonavir-boosted tipranavir.
a
Individual codons were counted, not multiple polymorphisms. Mixture of wild-type and mutant are counted as mutants. V3I was not counted.
b
No evidence of resistance detected using the HIV-1 genotyping method, versions 6.0 and 7.0 (TruGene).
c
Possible resistance detected using the HIV-1 genotyping method, versions 6.0 and 7.0 (TruGene).
d
Resistance detected using the HIV-1 genotyping method, versions 6.0 and 7.0 (TruGene).
e
Found to be susceptible or possibly resistant (excluding the study PI) by the HIV-1 genotyping method, versions 6.0 and 7.0 (TruGene);
enfuvirtide was always considered to be susceptible.
(TPV/r group, 1.6%; CPI/r group, 1.0%), and pneumonia
(TPV/r group, 1.6%; CPI/r group, 0.6%) were the most com-
mon adverse events. Severe adverse events (grade 3 or 4 ac-
cording to the Division of AIDS scoring system) were reported
for 71 patients (22.8%) in the TPV/r arm and 56 patients
(18.1%) in the CPI/r arm.
During the study, 15 types of AIDS-defining illnesses were
acquired by 11 patients (3.5%) in the TPV/r group and 16
patients (5.2%) in the CPI/r group. There was a higher fre-
quency of esophageal candidiasis in the CPI/r group (2.3% vs.
0.3%) and of wasting syndrome in the TPV/r group (1.0% vs.
0%). No other AIDS-defining illnesses were experienced by
12
patients in either group.
Laboratory abnormalities were generally more common in
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
1342 CID 2006:43 (15 November) HIV/AIDS
Figure 2. Comparison of virologic and immunologic responses in the ritonavir-boosted tipranavir (TPV/r) and ritonavir-boosted comparator protease
inhibitor (CPI/r) groups over 24 weeks. A, Treatment response (defined as a confirmed 1-log
10
reduction in the HIV-1 load). B, HIV-1 load reduction.
C, Virologic response (defined as an HIV-1 load
!50 copies/mL). D, CD4
+
cell count. For the TPV/r group, all 311 patients were analyzed for all variables
except for the CD4
+
cell count, for which 1 data point was missing ( ). All 309 patients in the CPI/r arm were analyzed for all 4 responsen p 310
variables.
the TPV/r arm than in the CPI/r arm (table 2); most of these
were mild or moderate and asymptomatic. Elevations in hepatic
enzyme and plasma lipid levels were the most common grade
3 or 4 laboratory abnormalities reported. Among patients who
experienced elevations in the ALT level, 15 (9.0%) were coin-
fected with hepatitis B or C virus, whereas 6 (4.4%) were not.
Among patients who experienced elevated aspartate amino-
transferase levels, 11 (6.6%) were coinfected with hepatitis B
or C virus, and 3 (2.2%) were not. Seventeen of 21 TPV/r
recipients with grade 3 or 4 liver function test results continued
to received treatment without permanent discontinuation.
No patient with grade 3 or 4 elevated ALT or AST levels in
either group developed treatment-onset hepatitis or related he-
patic events. Some patients were concomitantly receiving po-
tentially hepatotoxic drugs (13 [54.2%] of the 24 TPV/r-treated
patients and 5 [62.5%] of the 8 CPI/r-treated patients with
elevated ALT or AST levels).
There were 8 deaths (2.6%) in the TPV/r arm and 6 deaths
(1.9%) in the CPI/r arm (6.0 and 5.2 deaths per 100 patient-
exposure-years, respectively). None were judged to be related
to treatment.
DISCUSSION
The results of this 24-week interim analysis demonstrate that,
when TPV/r is used as part of antiretroviral combination ther-
apy, it significantly suppresses viral replication and increases
CD4
+
cell counts in genotypically screened, highly antiretro-
viral-treatment experienced patients with multidrug-resistant
HIV-1 infection. A significantly greater proportion of patients
randomized to receive TPV/r (41.5%), compared with CPI/r
(22.3%), achieved an HIV-1 load reduction of 1log
10
after
24 weeks. In addition, patients in the TPV/r group experienced
a significantly greater increase in CD4
+
cell counts than did
those in the CPI/r group, although small imbalances in baseline
characteristics may impact these results. These findings are im-
portant, because changes of this magnitude in virologic and
immunologic markers have been associated with a decreased
incidence of AIDS-defining events [29–32]. Furthermore, the
level of suppression of HIV-1 replication observed at 24 weeks
was comparable with that reported in the T-20 versus Opti-
mized Regimen Only (TORO) trials of treatment-experienced
patients, in which patients were randomized to receive enfu-
virtide or an optimized background regimen alone [33, 34].
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
HIV/AIDS CID 2006:43 (15 November) 1343
Table 2. Patients with a treatment response at week 24, according to baseline HIV-1
load.
Characteristic
Treatment group, no. of responders/
no. of evaluable patients (%)
Overall population TPV/r arm CPI/r arm
Total treated patients 198/620 (31.9) 129/311 (41.5) 69/309 (22.3)
Baseline HIV RNA level, copies/mL
!1000 1/4 (25.0) 1/3 (33.3) 0/1 (0)
1000–10,000 40/93 (43.0) 24/45 (53.3) 16/48 (33.3)
110,000 to 100,000 87/265 (32.8) 58/134 (43.3) 29/131 (22.1)
1100,000 70/258 (27.1) 46/129 (35.7) 24/129 (18.6)
NOTE. CPI/r, ritonavir-boosted comparator protease inhibitor; TPV/r, ritonavir-boosted tipranavir.
One treatment aim for patients with prior antiretroviral ex-
posure and drug resistance is to reestablish maximum virologic
suppression, and current guidelines recommend the addition
of a newer PI boosted with ritonavir, with or without enfu-
virtide, to a standard treatment regimen [35]. The results of
the RESIST-1 trial demonstrated that increasing the number of
background antiretroviral agents to which patients’ isolates
were susceptible enhanced the treatment response among both
TPV/r recipients and CPI/r recipients. This was true for the
TPV/r group when enfuvirtide, for which no level of cross-
resistance was expected, was added; the proportion of treatment
responders also increased (but less so) in the CPI/r arm. For
TPV/r recipients who were enfuvirtide naive, the inclusion of
enfuvirtide further enhanced the treatment response, compared
with that of patients who had a history of prior enfuvirtide
use. Similarly, having additional drugs beyond enfuvirtide in
the optimized background regimen to which patients’ isolates
were susceptible increased the proportion of treatment
responders.
Overall tolerability and the occurrence of adverse events were
comparable between treatment arms. Most adverse events were
mild or moderate in severity and were usually observed after
administration of boosted PIs in a population with advanced
immunodeficiency [36, 37]. Diarrhea, nausea, and fatigue were
the most frequently occurring adverse events reported in both
groups.
The grade 3 or 4 laboratory abnormalities observed in both
treatment groups were similar, although some occurred more
frequently in the TPV/r arm; asymptomatic elevations in ALT
and AST levels and increases in triglyceride and cholesterol
levels were more common among TPV/r recipients. Despite
these abnormalities, most patients continued to receive TPV/r
therapy. As seen with other boosted PIs, coinfection with hep-
atitis B or C virus and elevated transaminase levels at baseline
increased the risk of elevated ALT or AST levels during treat-
ment. Moreover, elevations in triglyceride levels were more
common among patients who had high triglyceride values at
baseline. One possible explanation for the observed increase in
liver events and in triglyceride levels is the higher total daily
dose of ritonavir used in TPV/r-treated patients (400 mg per
day). Average ritonavir trough concentrations are lower in pa-
tients receiving TPV/r than in those who receive some other
PIs (e.g., lopinavir and saquinavir) plus an optimized back-
ground regimen [38]; therefore, a higher daily dose of ritonavir
was used in tipranavir-treated patients in this study.
Because the trial was open label, investigators may have been
more prone to report adverse events for the investigational drug
than for approved agents [39]. More importantly, CPI/r-treated
patients were given the option of discontinuing treatment after
8 weeks in the event of a lack of initial virologic response or
confirmed virologic failure, to join a TPV/r rollover study. Be-
fore week 24, a total of 33.0% of CPI/r recipients opted to
discontinue receiving the study medication for these reasons,
and most of these patients joined the rollover study. This led
to a substantially longer treatment exposure among TPV/r re-
cipients than among CPI/r recipients. This greater exposure
may have led to an accumulation of adverse events (related or
unrelated to the study drug) in the TPV/r arm.
In conclusion, the combination of TPV/r with an active op-
timized background regimen in antiretroviral-experienced pa-
tients resulted in significant improvements in virologic and
immunologic responses through 24 weeks, compared with CPI/
r and an optimized background regimen. This would suggest
that TPV/r (500 mg/200 mg twice per day), as part of com-
bination antiretroviral therapy, plays an important role in the
achievement of effective viral suppression in patients infected
with multidrug-resistant strains who have limited treatment
options. The results of the 48-week analysis will determine the
durability of TPV/r in achieving and maintaining effective viral
suppression in this important patient population.
RESIST-1 STUDY GROUP
The resistance panel consisted of J. Baxter (Cooper Hospital,
Camden, NJ), C. A. Boucher (University of Utrecht, Utrecht,
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
1344 CID 2006:43 (15 November) HIV/AIDS
Table 3. Adverse events (related or not related to treatment; grade 1–4) and laboratory
abnormalities (grade 3–4 ) noted among study subjects.
Characteristic
Treatment group
Overall population
(n p 620)
TPV/r arm
(n p 311)
CPI/r arm
(n p 309)
Adverse event
Any
a
549 (88.5) 282 (90.7) 267 (86.4)
Diarrhea 144 (23.2) 75 (24.1) 69 (22.3)
Nausea 131 (21.1) 64 (20.6) 67 (21.7)
Fatigue 96 (15.5) 47 (15.1) 49 (15.9)
Headache 65 (10.5) 37 (11.9) 28 (9.1)
Vomiting 57 (9.2) 32 (10.3) 25 (8.1)
Pyrexia 53 (8.5) 29 (9.3) 24 (7.8)
Upper respiratory tract infection 45 (7.3) 24 (7.7) 21 (6.8)
Injection site reaction 51 (8.2) 22 (7.1) 29 (9.4)
Grade 3 or 4 laboratory abnormalities
Total no. of subjects 608 304 304
Elevated alanine aminotransferase level 25 (4.1) 21 (6.9) 4 (1.3)
Elevated aspartate aminotransferase level 19 (3.1) 14 (4.6) 5 (1.6)
Elevated amylase level 43 (7.0) 21 (6.9) 22 (7.2)
Elevated lipase level 14 (2.3) 9 (2.9) 5 (1.6)
Elevated cholesterol level 13 (2.1) 13 (4.2) 0
Elevated triglyceride level 104 (17.1) 66 (21.7) 38 (12.5)
Increased glucose level 11 (1.8) 6 (2.0) 5 (1.6)
NOTE. Data are no. (%) of subjects. CPI/r, ritonavir-boosted comparator protease inhibitor; TPV/r, rito-
navir-boosted tipranavir.
a
Averse events were observed in 15% of patients and were treatment related or not treatment related.
The Netherlands), and J. M. Schapiro (Stanford University,
Stanford, CA).
In addition to the listed authors, the RESIST-1 study group
included the following institutions and persons: B. Akil (Health
Innovations Research, Los Angeles, CA); M. Goldman, H. Kat-
ner, and F. C. Smail (University of Tennessee at Memphis); D.
Barker (CORE Center, Chicago, IL); W. Mazur (Early Inter-
vention Program [EIP] Clinic, Camden, NJ); S. Becker (Pacific
Horizon Medical Group, San Francisco, CA); K. Peterson (Na-
tional Naval Medical Center, Bethesda, MD); G. Blick (Circle
Medical, LLC, Norwalk, CT); C. Borkert (East Bay AIDS Center,
Berkeley, CA); A. Burnside (Burnside Clinic, Columbia, SC);
P. Cimoch (Orange County Center for Special Immunology,
Fountain Valley, CA); A. Collier (University of Washington
Harborview Medical Center, Seattle); E. DeJesus (IDC Research
Initiative, Altamonte Springs, FL); R. Eng (Veteran’s Affairs
New Jersey Health Care System, East Orange, NJ); J. Ernst
(AIDS Community Research Initiative of America [ACRIA],
New York); C. Farthing (AHF Research Center, Los Angeles);
J. Feinberg (University of Cincinnati Medical Center, Cincin-
nati, OH); J. Fessel (San Antonio Infectious Diseases Consult-
ants, San Antonio, TX); I. Frank (University of Pennsylvania
Medical Center, Division of Infectious Disease, Philadelphia);
M. Frank (Froedtert Memorial Lutheran Hospital, Milwaukee,
WI); J. Gallant (John Hopkins University School of Medicine,
Baltimore, MD); J. Gathe (Therapeutic Concepts, Houston,
TX); M. Goldman (Indiana University Hospital, Indianapolis);
R. Greenberg (University of Kentucky Medical Center, Lexing-
ton); P. Greiger-Zanlungo (Mount Vernon Hospital, Mount
Vernon, NY); B. Gripshove (University Hospitals of Cleveland,
Cleveland, OH); H. Grossman (Pollari Medical Group, New
York); T. Hawkins (Southwest CARE Center, Sante Fe, NM);
J. Hellinger (Community Research Initiative of New England,
Boston, MA); C. Hicks (Duke University Medical Center, Dur-
ham, NC); H. Horowitz (Westchester Medical Center, Valhalla,
NY); A. Huang (University of Louisville, Louisville, KY); D.
Jayaweera (Jackson Medical Tower, Miami, FL); J. Jemsek (Jem-
sek Clinic, Huntersville, NC); H. Katner (Mercer University
School of Medicine, Macon, GA); J. Fraiz (Infectious Disease
of Indiana, Indianapolis); H. Kessler (Rush-Presbyterian-St.
Luke’s Medical Center, Chicago); J. Kostman (Cecile Gallo,
Philadelphia FIGHT, Philadelphia, PA); A. Labriola (Washing-
ton VAMC, Washington); H. Lampiris (San Francisco VA Med-
ical Center, San Francisco); R. MacArthur (University Health
Center, San Francisco); D. Margolis (Dallas VAMC, Dallas); N.
Markowitz (Henry Ford Hospital, Detroit, MI); D. Mildvan
(Beth Israel Medical Center, New York); S. Miles (Clinical Re-
search Puerto Rico, Santurce, PR); A. Morris (Community Re-
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
HIV/AIDS CID 2006:43 (15 November) 1345
search Initiative of New England, Springfield, MA); R. A. Myers
(Phoenix Body Positive, Phoenix, AZ); J. Nadler (Hillsborough
County Health Dept., Tampa, FL); G. Pierone (Treasure Coast
Infectious Disease Consultants, Vero Beach, FL); G. Drusano
(Albany Medical College, Albany, NY); D. Rimland (Atlanta
VA Medical Ctr, Decatur, GA); M. Rodriguez (Houston Vet-
eran’s Administration, Houston); S. Santiago (CARES Re-
source, Miami); S. Schneider (Living Hope Clinical Trials, Long
Beach, CA); R. Schwartz (Associates In Research, Fort Myers,
FL); D. Norris (Comprehensive Research, Tampa); M. Sension
(North Broward Hospital District, Fort Lauderdale, FL); L. Sla-
ter (Infect Disease Inst. Clinical Trials Unit, Oklahoma City);
R. Smith (AIDS Consortium Service, Portland, ME); K. Squires
(University of Southern California/LA County USC Medical
Center, Los Angeles); R. Steigbigel (University of New York at
Stony Brook, Stony Brook); C. Steinhart (Steinhart Medical
Associates, Miami); R. Groger (Washington University AIDS
Clinical Trial Unit, St. Louis, MO); M. Thompson (AIDS Re-
search Consortium of Atlanta, Atlanta); F. Torriani (University
of California, San Diego, San Diego); V. Vega (Infectious Dis-
ease Associates, Sarasota, FL); D. Wheeler (Infectious Disease
Physicians Research, Annandale, VA); M. Wohlfeiler (Wohl-
feiler, Piperato & King, MD, Miami); B. Yangco (Infectious
Disease Research Institute, Tampa); J. Zachary (HIV Outpatient
Program [HOP], New Orleans, LO); T. Wilkin (The Cornell
HIV Clinical Trials Unit, New York); W. D. Hardy (Cedars-
Sinai Medical Center, Los Angeles); M. Wallace (Naval Medical
Center, San Diego, CA); D. Kuritzkes (Brigham Women’s Hos-
pital, Boston); R. Gandhi (Massachusettes General Hospital,
Boston); D. Ward (Dupont Circle Physicians Group, Washing-
ton); D. Berger (Northstar Medical, Chicago); D. Brand (North
Texas Center for AIDS and Clinical Research, Dallas); R. Corales
(Community Health Network, Rochester); T. File (Summa
Health System, HIV C.A.R.E. Center, Akron, OH); L. Tkatch
(Pinnacle Health, Harrisburg, PA); P. Bellman (The Office of
Dr. Paul Bellman, New York); H. Albrecht (Infectious Diseases
Clinics of Emory, Atlanta); D. Cooper (St. Vincents Hospital,
Darlinghurst, NSW, Australia); J. Gold (Albion Street Clinic,
Surry Hills, New South Wales [NSW]); J. Hoy (Alfred Hospital,
Melbourne, Victoria, Australia); C. Workman (Ground Zero,
Darlinghurst, NSW); J. Chuah (Gold Coast Sexual Health
Clinic, Miami, Queensland); M. Bloch (Holdsworth House
General Practice, Darlinghurst, NSW); D. Baker; W Cameron
(The Ottawa Hospital, Ottawa); B. Conway (Downtown In-
fectious Diseases Clinic, Vancouver, BC); P. Cote (Clinique
Medicale Du Quartier Latin, Montreal, QC); F. Crouzat (Ca-
nadian Immunodeficiency Research Collaborative Inc., To-
ronto, ON); K. Gough (St. Michael’s Hospital, Toronto, ON);
R Lalonde (Montreal Chest Institute, Montreal); A. Rachlis
(Sunnybrook & Women’s College Health Centre, Toronto, ON);
S. Rosser (St. Boniface General Hospital, Winnipeg, MB); F.
Smaill (McMaster University Medical Centre, Hamilton, ON);
B. Trottier (Clinique medicale l’Actuel, Montreal); S. Walmsley
(Toronto General Hospital, Toronto); C. Tsoukas (Montreal
General Hospital, McGill University Health Centre, Montreal);
L. Johnston (Centre For Clinical Research, Halifax, NOVA
SCO)
Acknowledgments
Financial support. Boehringer Ingelheim Pharmaceuticals.
Potential conflicts of interest. D.A.C. is a member of the speakers’
bureau for Boehringer Ingelheim GmbH. C.F. has received research study
grants from Boehringer Ingelheim and Abbott and is a member of the
speakers’ bureau and an advisory board participant for Boehringer Ingel-
heim and Abbott. D.J. is a consultant and member of the speakers’ bureau
for GlaxoSmithKline, Bristol-Myers Squibb, Roche, Johnson & Johnson,
Abbott, Boehringer Ingelheim GmbH, Virco, and Gilead and has received
research grants from GlaxoSmithKline, Bristol-Myers Squibb, Johnson &
Johnson, and Roche. G.P. has received research and educational grants
from Boehringer Ingelheim and is a member of the speakers’ bureau for
Boehringer Ingelheim GmbH. C.R.S. has served as an advisor for Boeh-
ringer Ingelheim. B.T. has been a member of the speakers’ bureau and an
advisory board participant for Boehringer Ingelheim GmbH. S.L.W. has
served as a consultant, on advisory boards, on speaker bureaus, and in the
conduct of clinical trials with Boehringer Ingelheim, Roche, Abbott, Bristol-
Myers Squibb GmbH, GlaxoSmithKline, Gilead, Tibotec, Merck, Pfizer,
and Agouron. C.W. has served as a consultant on advisory boards for Merck
Sharp & Dohme, Abbott, Roche, and Jansen-Cilag; has received honoraria
from Abbott, Roche, Jansen-Cilag, Gilead, and Merck Sharp & Dohme;
and has received grants from Abbott, Merck Sharp & Dohme, Roche,
Jansen-Cilag, Gilead, Bristol-Myers Squibb, GlaxoSmithKline, and Novar-
tis. G.M., V.K., C.D., S.M., D.M., and D.N. are all employees of Boehringer
Ingelheim. J.G. and D.N.: no conflicts.
References
1. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity
and mortality among patients with advanced human immunodefi-
ciency virus infection. HIV Outpatient Study Investigators. N Engl J
Med 1998; 338:853–60.
2. Cingolani A, Antinori A, Rizzo MG, et al. Usefulness of monitoring
HIV drug resistance and adherence in individuals failing highly active
antiretroviral therapy: a randomized study (ARGENTA). AIDS 2002;
16:369–79.
3. Wit FW, van Leeuwen R, Weverling GJ, et al. Outcome and predictors
of failure of highly active antiretroviral therapy: one-year follow-up of
a cohort of human immunodeficiency virus type 1-infected persons.
J Infect Dis 1999; 179:790–8.
4. Deeks SG. Determinants of virological response to antiretroviral ther-
apy: implications for long-term strategies. Clin Infect Dis 2000;
30(Suppl 2):S177–84.
5. Ledergerber B, Egger M, Opravil M, et al. Clinical progression and
virological failure on highly active antiretroviral therapy in HIV-1 pa-
tients: a prospective cohort study. Swiss HIV Cohort Study. Lancet
1999; 353:863–8.
6. Dieleman JP, Jambroes M, Gyssens IC, et al. Determinants of recurrent
toxicity-driven switches of highly active antiretroviral therapy. The
ATHENA cohort. AIDS 2002; 16:737–45.
7. Raboud JM, Harris M, Rae S, Montaner JS. Impact of adherence on
duration of virological suppression among patients receiving combi-
nation antiretroviral therapy. HIV Med 2002; 3:118–24.
8. Condra JH, Petropoulos CJ, Ziermann R, Schleif WA, Shivaprakash
M, Emini EA. Drug resistance and predicted virologic responses to
human immunodeficiency virus type 1 protease inhibitor therapy. J
Infect Dis 2000; 182:758–65.
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
1346 CID 2006:43 (15 November) HIV/AIDS
9. Hertogs K, Bloor S, Kemp SD, et al. Phenotypic and genotypic analysis
of clinical HIV-1 isolates reveals extensive protease inhibitor cross-
resistance: a survey of over 6000 samples. AIDS 2000; 14:1203–10.
10. Van Vaerenbergh K, Van Laethem K, Albert J, et al. Prevalence and
characteristics of multinucleoside-resistant human immunodeficiency
virus type 1 among European patients receiving combinations of nu-
cleoside analogues. Antimicrob Agents Chemother 2000; 44:2109–17.
11. Johnson VA, Brun-Vezinet F, Clotet B, et al. Drug resistance mutations
in HIV-1. Top HIV Med 2003; 11:215–21.
12. Lundgren JD, Ledergerber B, Fusco GP, et al. Risk of death following
triple class virological failure: the PLATO collaboration [abstract H-
450]. In: Program and abstracts of the 43rd Interscience Conference
on Antimicrobial Agents and Chemotherapy (Chicago). Washington,
DC: American Society for Microbiology, 2003.
13. Chrusciel RA, Strohbach JW. Non-peptidic HIV protease inhibitors.
Curr Top Med Chem 2004; 4:1097–114.
14. Mehandru S, Markowitz M. Tipranavir: a novel non-peptidic protease
inhibitor for the treatment of HIV infection. Expert Opin Investig
Drugs 2003; 12:1821–8.
15. Randolph JT, DeGoey DA. Peptidomimetic inhibitors of HIV protease.
Curr Top Med Chem 2004; 4:1079–95.
16. Larder BA, Hertogs K, Bloor S, et al. Tipranavir inhibits broadly pro-
tease inhibitor-resistant HIV-1 clinical samples. AIDS 2000; 14:1943–8.
17. Poppe SM, Slade DE, Chong KT, et al. Antiviral activity of the dihy-
dropyrone PNU-140690, a new nonpeptidic human immunodeficiency
virus protease inhibitor. Antimicrob Agents Chemother 1997; 41:
1058–63.
18. Back NK, van Wijk A, Remmerswaal D, et al. In-vitro tipranavir sus-
ceptibility of HIV-1 isolates with reduced susceptibility to other pro-
tease inhibitors. AIDS 2000; 14:101–2.
19. Khaliq Y, Gallicano K, Tisdale C, Carignan G, Cooper C, McCarthy
A. Pharmacokinetic interaction between mefloquine and ritonavir in
healthy volunteers. Br J Clin Pharmacol 2001; 51:591–600.
20. Koudriakova T, Iatsimirskaia E, Utkin I, Gangl E, Vouros P, Storozhuk
E. Metabolism of the human immunodeficiency virus protease inhib-
itors indinavir and ritonavir by human intestinal microsomes and ex-
pressed cytochrome P4503A4/3A5: mechanism-based inactivation of
cytochrome P4503A by ritonavir. Drug Metab Dispos 1998; 26:552–61.
21. Kumar GN, Dykstra J, Roberts EM, Jayanti VK, Hickman D. Potent
inhibition of the cytochrome P-450 3A-mediated human liver micro-
somal metabolism of a novel HIV protease inhibitor by ritonavir: a
positive drug-drug interaction. Drug Metab Dispos 1999; 27:902–8.
22. Tredger JM, Stoll S. Cytochromes P450-their impact on drug devel-
opment. Hospital Pharmacist 2002; 9:167–73.
23. MacGregor TR, Sabo JP, Norris SH, Johnson P, Galitz L, McCallister
S. Pharmacokinetic characterization of different dose combinations of
coadministered tipranavir and ritonavir in healthy volunteers. HIV Clin
Trials 2004; 5:371–82.
24. McCallister S, Valdez H, Curry K, et al. A 14-day dose-response study
of the efficacy, safety, and pharmacokinetics of the nonpeptidic protease
inhibitor tipranavir in treatment-naive HIV-1-infected patients. J Ac-
quir Immune Defic Syndr 2004; 35:376–82.
25. Cooper D, Hicks C, Cahn P, et al. 24-week RESIST study analyses: the
efficacy of tipranavir/ritonavir is superior to lopinavir/ritonavir, and
the TPV/r treatment response is enhanced by inclusion of genotypically
active antiretrovirals in the optimized background regimen [abstract
560]. In: Program and abstracts of the 12th Conference on Retroviruses
and Opportunistic Infections (Denver). Alexandria, VA: Foundation
for Retrovirology and Human Health, 2006.
26. Cahn P, Villacian J, Lazzarin A, et al. Ritonavir-boosted tipranavir
demonstrates superior efficacy to ritonavir-boosted protease inhibitors
in treatment-experienced HIV-infected patients: 24-week results of the
RESIST-2 trial. Clin Infect Dis 2006; 43:1347–56 (in this issue).
27. Ledergerber B, Lundgren JD, Walker AS, et al. Predictors of trend in
CD4-positive T-cell count and mortality among HIV-1-infected in-
dividuals with virological failure to all three antiretroviral-drug classes.
Lancet 2004; 364:51–62.
28. Cochran WG. Some methods for strengthening the common x
2
tests.
Biometrics 1954; 10:417–51.
29. O’Brien WA, Hartigan PM, Daar ES, Simberkoff MS, Hamilton JD.
Changes in plasma HIV RNA levels and CD4
+
lymphocyte counts
predict both response to antiretroviral therapy and therapeutic failure.
VA Cooperative Study Group on AIDS. Ann Intern Med 1997; 126:
939–45.
30. Mellors JW, Rinaldo CR Jr, Gupta P, White RM, Todd JA, Kingsley
LA. Prognosis in HIV-1 infection predicted by the quantity of virus
in plasma. Science 1996; 272:1167–70.
31. Human immunodeficiency virus type 1 RNA level and CD4 count as
prognostic markers and surrogate end points: a meta-analysis. HIV
Surrogate Marker Collaborative Group. AIDS Res Hum Retroviruses
2000; 16:1123–33.
32. Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-
controlled trial of ritonavir in advanced HIV-1 disease. The Advanced
HIV Disease Ritonavir Study Group. Lancet 1998; 351:543–9.
33. Lalezari JP, Henry K, O’Hearn M, et al. Enfuvirtide, an HIV-1 fusion
inhibitor, for drug-resistant HIV infection in North and South America.
N Engl J Med 2003; 348:2175–85.
34. Lazzarin A, Clotet B, Cooper D, et al. Efficacy of enfuvirtide in patients
infected with drug-resistant HIV-1 in Europe and Australia. N Engl J
Med 2003; 348:2186–95.
35. DHHS Panel on Antiretroviral Guidelines for Adults and Adoles-
cents–A Working Group of the Office of AIDS Research Advisory
Council (OARAC). Guidelines for the use of antiretroviral agents in
HIV-1-infected adults and adolescents. Available at: http://aidsinfo
.nih.gov/ContentFiles/AdultandAdolescentGL.pdf. Accessed 26 May
2006.
36. Murphy RL, Brun S, Hicks C, et al. ABT-378/ritonavir plus stavudine
and lamivudine for the treatment of antiretroviral-naive adults with
HIV-1 infection: 48-week results. AIDS 2001; 15:F1–9.
37. Pulido F, Katlama C, Marquez M, et al. A randomized study investi-
gating the efficacy and safety of amprenavir in combination with low-
dose ritonavir in protease inhibitor-experienced HIV-infected adults.
HIV Med 2004; 5:296–302.
38. Sabo JP, Piliero PJ, Lawton A, MacGregor TR, Leith J. A comparison
of steady-state trough plasma ritonavir concentrations for HIV+ pa-
tients receiving an optimized background regimen and ritonavir-
boosted tipranavir (TPV/r), lopinavir (LPV/r), saquinavir (SQV/r) or
amprenavir (APV/r) [poster 43]. In: Proceedings of the 7th Interna-
tional Workshop on Clinical Pharmacology of HIV Therapy (Lisbon).
Utrecht, The Netherlands: Virology Education, 2006.
39. Moore N, Hall G, Sturkenboom M, Mann R, Lagnaoui R, Begaud B.
Biases affecting the proportional reporting ratio (PPR) in spontaneous
reports pharmacovigilance databases: the example of sertindole. Phar-
macoepidemiol Drug Saf 2003; 12:271–81.
by guest on December 28, 2015http://cid.oxfordjournals.org/Downloaded from
... The new ARV + OBT approach vs placebo was first proposed by the TORO clinical trial [15]. Since then, this new ARV + OBT approach has been the most used rationale for evaluating new drugs for individuals with triple-class virological resistance [15][16][17][18][19]. However, the combinations studied have often not allowed direct comparisons between ARVs and have thus resulted in little evidence not only as to which the best combinations of two or three drugs are, but also as to which drug with a novel mechanism is to be chosen. ...
... Eighteen randomized controlled trials (totaling 7963 patients) comprising 47 ART comparison groups were retrieved [15][16][17][18][19][27][28][29][30][31][32][33][34][35][36][37][38][39] (Fig. 1 and Table 1). We also identified another 10 publications reporting extension results (referring to the previously selected RCTs that presented results in more advanced follow-up periods in time, i.e., 48 and 96 weeks) and 15 studies reporting subgroup analyses (Additional file 3: Table S2). ...
... The 18 RCTs assessed the efficacy and safety of nine new ARVs:ENF [15,39], TPV [16,28], DAPD [36], DRV [17,29,37], ETV [30][31][32], RAL [18,38], MVC [19,33], VIC [27,34], and DTG [35] (Table 1). Eleven of those studies (61%) were characterized as phase III trials and seven as phase IIb studies. ...
Article
Full-text available
Background The World Health Organization (WHO) has identified the need for evidence on third-line antiretroviral therapy (ART) for adults living with HIV/AIDS, given that some controversy remains as to the best combinations of ART for experienced HIV-1-infected patients. Therefore, we conducted a systematic review and meta-analysis to (i) assess the efficacy of third-line therapy for adults with HIV/AIDS based on randomized controlled trials (RCT) that adopted the “new antiretroviral (ARV) + optimized background therapy (OBT)” approach and (ii) address the key issues identified in WHO’s guidelines on the use of third-line therapy. Methods MEDLINE, EMBASE, LILACS, ISI Web of Science, SCOPUS, and Cochrane Central Register of Controlled Trials were searched for RCTs assessing third-line ARV therapy that used an OBT approach between 1966 and 2015. Data was extracted using an Excel-structured datasheet based on the Consolidated Standards of Reporting Trials (CONSORT) recommendations. The primary outcome of this meta-analysis was the proportion of patients reaching undetectable HIV RNA levels (< 50 copies/mL) at 48 weeks of follow-up. Included studies were evaluated using the Cochrane’s Risk of Bias assessment tool. Summarized evidence was rated according to the GRADE approach. Results Eighteen trials assessing 9 new ARV + OBT combinations defined as third-line HIV therapy provided the efficacy data: 7 phase IIb trials and 11 phase III trials. Four of the 18 trials provided extension data, thus resulting in 14 trials providing 48-week efficacy data. In the meta-analysis, considering the outcome regarding the proportion of patients with a viral load below 50 copies/ml at 48 weeks, 9 out of 14 trials demonstrated the superiority of the new combination being studied (risk difference = 0.18, 95% CI 0.13–0.23). The same analysis stratified by the number of fully active ARVs demonstrated a risk difference of 0.29 (95% CI 0.12–0.46), 0.28 (95% CI 0.17–0.38) and 0.17 (95% CI 0.10–0.24) respectively from zero, one, and two or more active drugs strata. Nine of the 18 trials were considered to have a high risk of bias. Conclusions Efficacy results demonstrated that the groups of HIV-experienced patients receiving the new ARV + OBT were more likely to achieve viral suppression when compared to the control groups. However, most of these trials may be at a high risk of bias. Thus, there is still not enough evidence to stipulate which combinations are the most effective for therapeutic regimens that are to be used sequentially due to documented multi-resistance.
... Interestingly, the ANRS algorithm showed that all tested samples were resistant to tipranavir/ritonavir (TPV/ r) drugs that are unavailable in Bangladesh. TPV/r, a protease inhibitor, was evaluated as an initial therapy for treatment-naive HIV-1-infected patients because of its potency, unique resistance profile, and high genetic barrier (Gathe et al., 2006). Therefore, the careful use of tipranavir/ritonavir (TPV/r) drugs for HIV treatment in Bangladesh is suggested. ...
Article
Full-text available
Objective: HIV-1 subtyping data of Bangladeshi strains are available in global HIV Sequence Database up to 2007, and there is no sequence of drug resistance profile based on the pol gene segment. This study aimed to update HIV genotyping data and describe the drug resistance mutations for the first time from Bangladesh using specimens from the latest HIV sero-surveillance conducted in 2016. Study design and methods: During HIV sero-surveillance, a total of 1268 people who inject drugs (PWID) and 3765 female sex workers (FSW) were screened and among them, 230 (18.1%) PWID and 7 (0.2%) FSW were HIV positive. Among HIV positives, randomly selected 74 specimens (60 male-PWID, 7 female-PWID, and 7 FSW) were subjected to gag, pol, and env gene sequencing using gene-specific primers. Genotyping was decided based on the partial gag and env genes while transmission dynamics was based on the gag sequence (n = 237). Drug resistance profiles were obtained by using the algorithm of the established available drug resistance database. Results: HIV subtype C and C-related recombinants have remained the major circulating genotypes in Bangladesh. Although the recurring transmission of subtype C occurred among PWID, we identified possible transmission to other key populations (KPs), which suggests spillover from PWID through the sexual route. The prevalence of drug-resistant mutation was low, and all strains were susceptible to NRTIs and NNRTIs drugs. Unique recombination forms (URF) with genotype C for gag-pol and A1 for env was also identified. Conclusions: The study findings warrant continuous monitoring of HIV-positive individuals and future investigation to identify social networks within and between KPs to halt the transmission and prevent new infections.
... Interestingly, the ANRS algorithm showed that all tested samples were resistant to tipranavir/ritonavir (TPV/ r) drugs that are unavailable in Bangladesh. TPV/r, a protease inhibitor, was evaluated as an initial therapy for treatment-naive HIV-1-infected patients because of its potency, unique resistance profile, and high genetic barrier (Gathe et al., 2006). Therefore, the careful use of tipranavir/ritonavir (TPV/r) drugs for HIV treatment in Bangladesh is suggested. ...
Preprint
Full-text available
Objective: HIV-1 subtyping data of Bangladeshi strains are available in global HIV Sequence Database up to 2007, and there is no sequence of drug resistance profile based on pol gene segment. This study aimed to update HIV genotyping data, and describe the drug resistance mutations for the first time from Bangladesh using specimens from the latest HIV sero-surveillance conducted in 2016. Study Design and methods: During HIV sero-surveillance, a total of 1,268 people who inject drugs (PWID) and 3,765 female sex workers (FSW) were screened among them, 230 (18.1%) PWID and 7 (0.2%) FSW were HIV positive. Among HIV positives, randomly selected 74 specimens (60 male-PWID, 7 female-PWID and 7 FSW) were subjected for gag, pol and env gene sequencing using gene-specific primers. Genotyping was decided based on the partial gag and env genes while transmission dynamics was based on gag sequence (n=237). Drug resistance profiles were obtained by using the algorithm of the established available drug resistance database. Results: HIV subtype C and C-related recombinants have remained the major circulating genotypes in Bangladesh. Although recurring transmission of subtype C occurred among PWID, we identified possible transmission to other key population suggesting spillover from PWID through the sexual route. The prevalence of drug-resistant mutation was low, and all strains were susceptible to NRTIs and NNRTIs drugs. Unique recombination forms (URF) with genotype C for gag-pol and A1 for env was also identified. Conclusions: The study findings warrant continuous monitoring of HIV positive individuals and future investigation to identify social networks within and between KPs to halt the transmission and prevent new infections.
... Adverse Cutaneous Reactions Associated with the Newest Antiretroviral Drugs • Tipranavir: a protease inhibitor (PI) approved for use with ritonavir for patients with PI resistance; the most common ACDR are urticaria, morbilliform rash or photosensitive lesions with onset usually around day 53, occurring in up to 8-14% of patients [55]; it contains a sulphonamide moiety, and sulpha-containing drugs should be used with caution, although sulphonamide allergy is not an absolute contraindication [8,56,57] • Darunavir: in clinical trials of this PI, rashes occurred in 7% of cases, which were mostly self-limited and morbilliform; 1 case of SJS was reported [8,58] • Etravirine: a new NNRTI which has been used in patients with NNRTI resistance; ACDR were reported in 16.9% of cases, the majority of which were mild-to-moderate morbilliform lesions arising in the first few weeks and resolving with continued treatment; the reactions were more common in females than in males; severe reactions including erythema multiforme, SJS and DHS were reported in < 0.1% of patients [8,59,60] • Raltegravir: an integrase inhibitor; patients in trials were reported to present with mildto-moderate rash and pruritus, with 2 cases of hypersensitivity [8,61] • Maraviroc: a CCR5 chemokine receptor inhibitor; a pruritic rash occurred in 3.8% of patients, which may precede hepatotoxicity; therefore, patients with rash, eosinophilia and raised IgE should be evaluated for hepatotoxicity [8,62] Hypersensitivity Syndrome This is a life-threatening reaction which occurs in the first 42 days of antiretroviral therapy. It presents with a diffuse maculopapular eruption, fever, eosinophilia, atypical lymphocytosis, multivisceral involvement and raised liver enzyme levels (> 5-times elevated transaminases). ...
Article
Full-text available
Background: The global mortality from HIV and the cutaneous burden of infective, inflammatory and malignant diseases in the setting of AIDS have significantly declined following the advent of highly active antiretroviral therapy. Regrettably, there has been a contemporaneous escalation in the incidence of adverse cutaneous drug reactions (ACDR), with studies attesting that HIV-positive individuals are a hundred times more susceptible to drug reactions than the general population, and advanced immunodeficiency portending an even greater risk. Several variables are accountable for this amplified risk in HIV. Summary: Adverse reactions to trimethoprim-sulfamethoxazole are the most common, increasing from approximately 2-8% in the general population over to 43% amongst HIV-positive individuals to approximately 69% in subjects with AIDS. Antituberculosis drugs and antiretrovirals are also well-known instigators of ACDR. Cutaneous reactions range from mild morbilliform eruptions to severe, life-threatening manifestations in the form of Stevens-Johnson syndrome/toxic epidermal necrolysis. Histological features vary from vacuolar interface changes to full-thickness epidermal necrosis with subepidermal blister formation. A precipitous diagnosis of the ACDR, clinically and histologically if necessary, together with the isolation of the causative drug is critical. The identification process, however, is often complex and multifaceted due to polypharmacy and inconclusive data on which drugs are the most likely offending agents, especially against the background of tuberculosis co-infection. Key messages: Whilst milder cutaneous reactions are treated symptomatically, severe reactions mandate immediate treatment discontinuation without rechallenge. Further studies are required to establish safe rechallenge guidelines in resource-limited settings with a high HIV and tuberculosis prevalence.
... Tipranavir has been available for treatment since 2005 and is commercialized in two oral dosage forms: a solution containing 100 mg mL −1 and a capsule containing 250 mg in a self-emulsifying drug delivery system (SEDDS), to improve its bioavailability [8]. It is used in a ritonavir combined therapy twice daily [2,9]. ...
Article
Tipranavir (TPV) is one of the most recently developed protease inhibitors (PI) and it is specially recommended for treatment-experienced patients who are resistant to other PI drugs. In this work, a simple and friendly environmental CZE stability-indicating method to assay TPV capsules was developed and two TPV organic impurities were identified by high resolution mass spectrometry (HRMS). The optimized analytical conditions were: background electrolyte composed of sodium borate 50 mM, pH 9.0 and 5% of methanol; voltage + 28 kV; hydrodynamic injection of 5 s (100 mbar), detection wavelength 240 nm, at 25 °C. The separation was achieved in a fused silica capillary with 50 µm × 40 cm (inner diameter × effective length), using furosemide as internal standard. All the validation parameters were met and the method was specific, even in the presence of degradation products and impurities. Oxidation was indicated as the main degradation pathway among those evaluated in this study (acidic, alkaline, thermal, photolytic and oxidative) and it showed a second order degradation kinetic, under the conditions used in this study. The main oxidation product and an organic impurity detected in the standard were characterized by Q-TOF, and both of them correspond to oxidation products of TPV.
Article
Heavily treatment-experienced (HTE) persons with HIV have limited options for antiretroviral therapy and face many challenges, complicating their disease management. There is an ongoing need for new antiretrovirals and treatment strategies for this population. We reviewed the study designs, baseline characteristics, and results of clinical trials that enrolled HTE persons with HIV. A PubMed literature search retrieved articles published between 1995 and 2020, which were grouped by trial start date (1995–2009, N = 89; 2010–2014, N = 3; 2015–2020, N = 2). Clinical trials in HTE participants markedly declined post-2010. Participant characteristics and study designs showed changes in trends over time. As treatment strategies for HTE persons with HIV progress, we must look beyond virologic suppression to consider the broader needs of this complex heterogeneous population.
Conference Paper
The introduction of antiretroviral therapy (ART) has transformed HIV from a fatal illness into a manageable, chronic condition. As there is currently no cure for HIV, ART, typically including three antiretroviral drugs (ARVs), is a lifelong commitment, and there are concerns around long-term toxicities. Integrase strand transfer inhibitors (INSTIs) are one of the latest ARV classes to be approved and treatment guidelines uniformly recommend them as first-line treatment for people living with HIV (PLWH). However, limited data exist on long-term clinical outcomes associated with contemporary ART, including INSTIs, and contemporary ART regimens, including two-drug regimens. In this thesis, I use data from the International Cohort Consortium of Infectious Diseases (RESPOND) to assess the use and outcomes of contemporary ART, with a focus on two-drug regimens and INSTIs, including the association between INSTI use and incident cancer. I also assess temporal trends in cancer incidence across different ART-eras. RESPOND is a collaboration of 17 cohort studies, including approximately 30,000 PLWH from across Europe and Australia. I found that uptake of dolutegravir compared to cobicistat-boosted elvitegravir or raltegravir has increased over time. INSTI discontinuation was low overall and mainly due to toxicity in the first 6 months of use. Discontinuation was higher for raltegravir, primarily due to treatment simplification, whilst discontinuation due to nervous system toxicities was highest on dolutegravir. Virological and immunological outcomes were similar between those on two-drug and three-drug regimens. Additionally, after accounting for baseline characteristics, there was a similar incidence of severe clinical events on both regimen types. When assessing cancer trends from 2006-2019, I found that whilst the age-standardised incidence of AIDS-related and infection-related cancers has decreased over time, body mass index-related cancers have increased, whilst non-AIDS-related cancers and smoking-related cancers remained constant. Overall, there was no association between cancer risk and INSTI exposure.
Article
Tipranavir (TPV) is a protease inhibitor (PI) specially recommended for treatment-experienced patients who are resistant to other PI drugs. It was approved by FDA in 2005, but, until now, it has not been included in any official compendia. The objective of this study is to develop and validate a simple LC–UV method to assay TPV capsules, and to study the cytotoxicity of TPV and degraded samples over the cell viability. The optimized conditions were C8 Phenomenex® column (Luna®, 150 mm × 4.6 mm, 5 µm); mobile phase composed by methanol, acetonitrile, and acidified water pH 3.5 (40:31:29); flow rate 1.0 mL min−1 and detection at 254 nm. The cytotoxic effects of non-degraded and degraded TPV samples were evaluated in 3T3 cells by means of MTT viability assay. The method was linear in the range of 10–100 µg mL−1 (r = 0.9999) and specific, even in the presence of degradation products and impurities. The method showed suitable accuracy (mean recovery 100.02%), precision (RSD < 1.10%), and a two-level full factorial design indicated the robustness of the method. The degraded samples exhibited cytotoxicity patterns similar to TPV under controlled conditions. The method developed is appropriate for quality control of TPV capsules and stability studies.
Chapter
HIV protease is pivotal in the viral replication cycle and directs the formation of mature infectious virus particles. The development of highly specific HIV protease inhibitors (PIs) , based on thorough understanding of the structure of HIV protease and its substrate, serves as a prime example of structure-based drug design. The introduction of first-generation PIs marked the start of combination antiretroviral therapy . However, low bioavailability, high pill burden, and toxicity ultimately reduced adherence and limited long-term viral inhibition. Therapy failure was often associated with multiple protease inhibitor resistance mutations, both in the viral protease and its substrate (HIV gag protein), displaying a broad spectrum of resistance mechanisms. Unfortunately, selection of protease inhibitor resistance mutations often resulted in cross-resistance to other PIs.
Article
BACKGROUND & METHODS: National surveillance data show recent, marked reductions in morbidity and mortality associated with the acquired immunodeficiency syndrome (AIDS). To evaluate these declines, we analyzed data on 1255 patients, each of whom had at least one CD4+ count below 100 cells/mm' who were seen at 9 clinics specializing in the treatment of human immunodeficiency virus (HIV) infection in 8 U.S. cities from January 1994 through June 1997. RESULTS: Mortality among the patients declined from 29.4/100 person-yrs in the first quarter of 1995 to 8.8/100 in the second quarter of 1997. There were reductions in mortality regardless of sex, race, age, and risk factors for transmission of HIV. The incidence of any of 3 major opportunistic infections (Pneumocystis curinii pneumonia, Mycohacterium avium complex disease, and cytomegalovirus retinitis) declined from 21.9 /100 person-yrs in 1994 to 3.7/100 person-yrs by mid-1997. In a failure-rate model, increases in the intensity of antiretroviral therapy (classified as none, monotherapy, combination therapy without a protease inhibitor, and combination therapy with a protease inhibitor) were associated with stepwise reductions in morbidity and mortality. Combination antiretroviral therapy was associated with the most benefit; the inclusion of protease inhibitors in such regimens conferred additional benefit. Patients with private insurance were more often prescribed protease inhibitors and had lower mortality rates than those insured by Medicare or Medicaid. CONCLUSIONS: The recent declines in morbidity and mortality due to AIDS are attributable to the use of more intensive antiretroviral therapies.
Article
Objective: To evaluate treatment-mediated changes in HIV-1 RNA and CD4 count as prognostic markers and surrogate end points for disease progression (AIDS/death). Methods: Data from 13,045 subjects in all 16 randomized trials comparing nucleoside analogue reverse transcriptase inhibitors and having HIV-1 RNA measurements at 24 weeks were obtained. A total of 3146 subjects had HIV-1 RNA and CD4 count determinations at 24 weeks after starting treatment. Results: At Week 24, the percentage of subjects experiencing an HIV-1 RNA decrease of >1 log10 copies/ml or a CD4 count increase of >33% was similar (22% vs 25%). Changes in both markers at Week 24 were significant independent predictors of AIDS/death: Across trials, the average reduction in hazard was 51% per 1 log10 HIV-1 RNA copies/ml decrease (95% confidence interval: 41%, 59%) and 20% per 33% CD4 count increase (17%, 24%). In univariate analyses, the hazard ratio for AIDS/death in randomized treatment comparisons was significantly associated with differences between treatments in mean area under the curve of HIV-1 RNA changes to Weeks 8 and 24 (AUCMB) and mean CD4 change at Week 24, but, in multivariate analysis, only mean CD4 change was significant. Conclusions: Change in HIV-1 RNA, particularly using AUCMB, and in CD4 count should be measured to aid patient management and evaluation of treatment activity in clinical trials. However, short-term changes in these markers are imperfect as surrogate end points for long-term clinical outcome because two randomized treatment comparisons may show similar differences between treatments in marker changes but not similar differences in progression to AIDS/death.
Article
In recent times, major advances have been made in understanding how drugs are metabolised. This article discusses the mechanisms involved, with particular attention paid to cytochrome P450 enzymes.
Article
Tipranavir (TPV), a novel nonpeptidic protease inhibitor (NPPI), was administered to treatment-naive HIV-1-infected patients over 14 days in a randomized, multicenter, open-label, parallel-group trial to evaluate the efficacy and tolerability of a self-emulsifying drug delivery system (SEDDS) formulation, in combination with ritonavir (RTV). Of the 31 patients enrolled, 10 were randomized to receive TPV 1200 mg twice daily (TPV 1200), 10 patients received TPV 300 mg + RTV 200 mg twice daily (TPV/r 300/200), and 11 patients received TPV 1200 mg + RTV 200 mg twice daily (TPV/r 1200/200). The median baseline viral load and CD4 cell count were 4.96 log(10) copies/mL and 244 cells/mm(3), respectively. After 14 days, the median decrease in viral load was -0.77 log(10) in the TPV 1200 group, -1.43 log(10) in the TPV/r 300/200 group, and -1.64 log(10) in the TPV/r 1200/200 group. TPV exposure was increased by 24- and 70-fold in the TPV/r 300/200 and 1200/200 groups, respectively, compared with TPV 1200 alone. There were no significant differences across treatment arms with regard to drug-related adverse events. TPV/r appeared to be safe, effective, and well tolerated during 14 days of treatment.
Article
Background Ritonavir is a potent, orally bioavailable inhibitor of HIV-1 protease. We undertook an international, multicentre, randomised, double-blind, placebo-controlled trial of ritonavir in patients with HIV-1 infection and CD4-lymphocyte counts of 100 cells/u, L or less, who had previously been treated with antiretroviral drugs. Methods 1090 patients were randomly assigned twice-daily liquid oral ritonavir 600 mg (n=543) or placebo (n=547) while continuing treatment with up to two licensed nucleoside agents. The primary study outcome was any first new, or specified recurrent, AIDS-defining event or death. Open-label ritonavir was provided after 16 weeks in the study to any patient who had an AIDS-defining event. Findings The baseline median CD4-lymphocyte count was 18 (IQR 10-43)/μ in the ritonavir group and 22 (10-47) /μL in the placebo group. Study medication was discontinued in 114 (21·1%) ritonavir-group patients and 45 (8·3%) placebo-group patients mainly because of initial adverse symptoms. Outcomes of AIDS-defining illness or death occurred in 119 (21·9%) ritonavir-group patients and 205 (37·5%) placebo-group patients (hazard ratio 0·53 [95% Cl 0·42-0·66]; log-rank p<0·0001) during median follow-up of 28·9 weeks, with loss to follow-up of 15 (1·4%) patients. Ritonavir was then offered to all patients; at median follow-up of 51 weeks, 87 (16%) ritonavir-group patients had died of any cause versus 126 (23%) placebo-group patients (hazard ratio 0·69 [95% Cl 0·52-0·91], log-rank p=0·0072). Interpretation Although earlier intervention with combination therapy may provide much more effective treatment, ritonavir in patients with advanced disease and extensive previous antiretroviral use is safe and effective, lowers the risk of AIDS complications, and prolongs survival.
Article
Objective: To draw attention to the many cases of Stevens-Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN) related to nevirapine detected in a multinational case-control study of SJS and TEN. Methods: Actively detected cases and matched hospital controls were interviewed for exposure to drugs and other risk factors. Data were analysed with case-control and case-crossover methods. Results: Between May 1997 and November 1999, a diagnosis of SJS or TEN was established in 246 patients. Eighteen were known to be infected by HIV-1 (7.3%), 15 out of these 18 had been exposed to nevirapine. The reaction began 10-240 days after the introduction of nevirapine (median, 12 days) and all patients had received escalating doses. In 10 patients the reaction occurred with the initial dosage. All but one patients received simultaneously a variety of other antiretroviral agents but no specific drug combination emerged, and nevirapine was the only drug significantly associated with an increased risk of SJS or TEN in HIV-infected persons [odds ratio, 62 (10.4; +∞) in the case-control analysis; odds ratio, +∞ (2.8; +∞) in the case-crossover analysis]. Conclusions: In European countries the risk of SJS or TEN in the context of HIV infection appears to be associated with nevirapine. The respect of a lead-in period does not appear to prevent SJS or TEN. Because of the severity of these reactions and the long elimination half-life of nevirapine, we suggest discontinuation of the drug as soon as any eruption occurs.
Article
Background: Acquired HIV-specific cell-mediated immune responses have been observed in exposed-uninfected individuals, and it has been inferred, but not demonstrated, that these responses constitute a part of natural protective immunity to HIV. This inference was tested prospectively in the natural exposure setting of maternal-infant HIV transmission in a predominantly breast-fed population. Methods: Cord blood from infants of HIV-seropositive women in Durban, South Africa, were tested for in vitro reactivity to a cocktail of HIV envelope peptides (Env) using a bioassay measuring interleukin-2 production in a murine cell line. Infants were followed with repeat HIV RNA tests up to 18 months of age to establish which ones acquired HIV-infection. Results: T-helper cell responses to Env were detected in 33 out of 86 (38%) cord blood samples from infants of HIV-seropositive women and in none of nine samples from seronegative women (P = 0.02). Among infants of HIV-seropositive mothers, three out of 33 with T-helper responses to Env were already infected before delivery (HIV RNA positive on the day of birth), two were lost to follow-up, and none of the others (out of 28) were found to be HIV infected on subsequent tests. In comparison, six out of 53 infants unresponsive to Env were infected before delivery, and eight out of 47 (17%) of the others were found to have acquired HIV infection intrapartum or post-partum through breast-feeding (P = 0.02). Conclusions: T-helper cell responses to HIV envelope peptides were detected in more than one-third of newborns of HIV-infected women; no new infections were acquired by these infants at the time of delivery or post-natally through breast-feeding if these T-helper cell responses were detected in cord blood.