ArticlePDF Available

A Pilot Study of Switch to Lopinavir/Ritonavir (LPV/r) Monotherapy from Nonnucleoside Reverse Transcriptase Inhibitor–Based Therapy

Authors:
  • AIDS Research and Treatment Center of the Treasure Coast

Abstract and Figures

This study evaluated the safety and efficacy of switching HIV-infected patients with stable viral suppression on nonnucleoside reverse transcriptase inhibitor/nucleoside reverse transcriptase inhibitor (NNRTI/NRTI) therapy to lopinavir/ritonavir (LPV/r) monotherapy. Eligible patients discontinued NNRTI and started LPV/r. Two weeks later NRTIs were stopped and LPV/r monotherapy was continued. Patients were seen every 4 weeks throughout the 48-week study. Twelve of 18 (66%) participants met the primary endpoint with HIV RNA <75 copies/mL at week 48. Thirteen (72%) participants completed 48 weeks of LPV/r monotherapy, and 12 of 13 (92%) participants on treatment at week 48 had HIV RNA <75 copies/mL. Ten (55%) of 18 patients maintained HIV RNA <75 copies/mL at all time points. Two patients were withdrawn with virologic failure but demonstrated no evidence of virologic resistance. Three (17%) patients withdrew due to diarrhea, 2 with hyperglycemia at baseline developed diabetes mellitus, 7 (54%) required addition of or increase in lipid-lowering agents, but none had grade 3 or 4 hyperlipidemia. Results from this pilot study suggest that LPV/r monotherapy may be an option for management of HIV infection. Larger, randomized trials are warranted to evaluate the safety, efficacy, and patient population who might benefit from LPV/r monotherapy.
Content may be subject to copyright.
237
A Pilot Study of Switch to Lopinavir/Ritonavir
(LPV/r) Monotherapy from Nonnucleoside Reverse
Transcriptase Inhibitor–Based Therapy
Gerald Pierone, Jr., MD,1,2 Jeffrey Mieras, RN,2 Dorothy Bulgin-Coleman, ARNP,1
Chandra Kantor, ARNP,1 James Shearer, PA,1 Lunie Fontaine, MT (ASCP),3
Michael Fath, PhD,3 and Michael Norton, PA3
1AIDS Research and Treatment Center of the Treasure Coast, Ft. Pierce, Florida;
2Treasure Coast Infectious Disease Consultants, Vero Beach, Florida;
3Abbott Laboratories, Abbott Park, Illinois, USA
For correspondence or reprints contact: Gerald Pierone, Jr., MD,
Treasure Coast Infectious Disease Consultants, 3715 7th Ter-
race, Vero Beach, FL 32960 USA. Email: pieroneg@bellsouth.net
HIV Clin Trials 2006;7(5):237–245
© 2006 Thomas Land Publishers, Inc.
www.thomasland.com
doi:10.1310/hct0705-237
Purpose: This study evaluated the safety and efficacy of switching HIV-infected
patients with stable viral suppression on nonnucleoside reverse transcriptase
inhibitor/nucleoside reverse transcriptase inhibitor (NNRTI/NRTI) therapy to
lopinavir/ritonavir (LPV/r) monotherapy. Method: Eligible patients discontinued
NNRTI and started LPV/r. Two weeks later NRTIs were stopped and LPV/r
monotherapy was continued. Patients were seen every 4 weeks throughout the 48-
week study. Results: Twelve of 18 (66%) participants met the primary endpoint with
HIV RNA <75 copies/mL at week 48. Thirteen (72%) participants completed 48
weeks of LPV/r monotherapy, and 12 of 13 (92%) participants on treatment at week
48 had HIV RNA <75 copies/mL. Ten (55%) of 18 patients maintained HIV RNA <75
copies/mL at all time points. Two patients were withdrawn with virologic failure but
demonstrated no evidence of virologic resistance. Three (17%) patients withdrew
due to diarrhea, 2 with hyperglycemia at baseline developed diabetes mellitus, 7
(54%) required addition of or increase in lipid-lowering agents, but none had grade 3
or 4 hyperlipidemia. Conclusion: Results from this pilot study suggest that LPV/r
monotherapy may be an option for management of HIV infection. Larger,
randomized trials are warranted to evaluate the safety, efficacy, and patient
population who might benefit from LPV/r monotherapy. Key words: HAART, highly
active antiretroviral therapy, lopinavir/ritonavir, LPV/r, NNRTIs, nonnucleoside
reverse transcriptase inhibitors, NRTIs, nucleoside reverse transcriptase inhibitors,
PIs, protease inhibitors
T
he US Department of Health and Human
Services (DHHS) guidelines for treatment of
HIV infection in adults and adolescents list
two preferred highly active antiretroviral therapy
(HAART) regimens. Both options comprise three
drugs, including two nucleoside or nucleotide
reverse transcriptase inhibitors (NRTIs), together
with either the protease inhibitor (PI) lopinavir/
ritonavir (LPV/r) or the nonnucleoside reverse
transcriptase inhibitor (NNRTI) efavirenz.1
Despite remarkable advances in management of
HIV2 since the advent of HAART, standard therapy
has several disadvantages. Clinically significant
side effects may be seen with all classes of
antiretroviral agents. NRTIs have been associated
with mitochondrial toxicity,1,3–5 which may mani-
fest as lactic acidosis, hepatic failure, neuropathy,
pancreatitis, and lipoatrophy. NNRTIs have been
associated with hepatotoxicity, hyperlipidemia,
and neuro-cognitive dysfunction. In addition, cur-
rently available NNRTIs demonstrate a low barrier
to the development of resistance, requiring the se-
lection of only a single mutant population to ren-
der loss of antiviral activity and cross-resistance to
other NNRTIs.6 PIs are associated with varying de-
grees of gastrointestinal side effects and metabolic
disturbances.1 Although the overall cost of care for
238 HIV CLINICAL TRIALS 7/5 SEPT-OCT 2006
adult HIV patients since the introduction of suc-
cessful combination therapy has declined, the an-
nual cost for the antiretrovirals that make up
HAART has increased.7 Hence, the identification of
an effective antiretroviral therapy that avoids long-
term exposure to three different agents and is less
expensive could be a useful option for the manage-
ment of HIV infection.
Earlier studies of HAART induction followed by
simplification to unboosted PI monotherapy were
unsuccessful.8–10 However, as a result of recent ob-
servations made with ritonavir (r)-boosted PI
therapy,11–14 an interest in the investigation of
boosted PIs as monotherapy for HIV treatment has
emerged.
LPV/r is currently the only co-formulated
boosted PI available. LPV/r is considered to be a
single antiretroviral agent because the levels of
ritonavir achieved in plasma are subinhibitory and
therefore the antiretroviral effect of the combina-
tion is based on the activity of lopinavir alone.15
When LPV/r is dosed twice daily, trough lopinavir
plasma levels are a median 84-fold higher than the
protein binding–adjusted 50% inhibitory concen-
tration (IC50) of lopinavir against wild-type HIV-
1.15 This high inhibitory quotient (IQ = trough
lopinavir concentration/IC50) may partially ex-
plain the clinical observation that primary PI resis-
tance has rarely been seen in patients who have
received LPV/r as part of their first HAART regi-
men.16,17 Due to the potency of LPV/r, the low fre-
quency of selection for resistance to LPV/r, and the
potential for cost savings, a 48-week pilot study of
switching to LPV/r monotherapy was conducted
and the results are reported herein.
METHOD
Study Design
This was a 48-week prospective pilot study in
which HIV-1–infected patients with viral suppres-
sion to <75 copies/mL on an NNRTI-based regi-
men were switched to LPV/r monotherapy. A
planned target of 15 patients was chosen to provide
preliminary data on the safety and efficacy of LPV/r
monotherapy. The study was approved by the
Western Institutional Review Board, and written
informed consent was obtained from each patient
prior to any study procedures being performed. At
baseline, patients were instructed to discontinue
the NNRTI and substitute LPV/r 400/100 mg soft
gel capsules (three capsules twice a day). NRTIs
were continued for 2 weeks to provide an overlap
of antiretroviral coverage in case LPV plasma lev-
els were excessively lowered from the known drug
interaction with NNRTIs. At day 14, the NRTIs
were stopped and LPV/r was continued as
monotherapy. Study visits were scheduled every 4
weeks throughout the 48-week study period. At
each visit, a self-administered adherence question-
naire was completed and pill count was per-
formed.18 Vital sign measurements, abbreviated
physical examination, and assessment for adverse
events (AEs) were also performed at each visit.
Concomitant medications including the use of
lipid-lowering agents (LLAs) were recorded. Tele-
phone contact from the study team to each patient
was attempted between study visits to monitor
changes in the patient’s status and to reinforce the
importance of adherence to treatment.
Blood samples for CD4+ lymphocyte counts,
HIV RNA levels, hematology, and chemistry test-
ing were collected every 4 weeks for the first 24
weeks and every 8 weeks thereafter. Fasting
plasma lipid levels were collected at baseline and
at weeks 8, 24, and 48. A plasma sample was stored
for each patient at weeks 2 and 24 for ascertaining
LPV levels in cases of virologic failure (VF). Addi-
tionally, whenever a patient was found to have
viral load >400 copies/mL, a blood sample was
scheduled for collection at the succeeding visit for
resistance testing.
HIV RNA levels were determined by bDNA
technology (Bayer Diagnostics, Tarrytown, New
York, USA). Genotyping was performed as needed
using a Virco HIV-1 Virtual Phenotype.
Phenotyping was performed using Virologic
Phenosense GT.
Inclusion/Exclusion Criteria
Participants were recruited from two affiliated
HIV treatment clinics in Florida: the AIDS Research
and Treatment Center of the Treasure Coast in Ft.
Pierce, and Treasure Coast Infectious Disease Con-
sultants in Vero Beach. Patients were eligible for
enrollment if they were over 18 years of age, naïve
to PIs, and on a stable NNRTI-based antiretroviral
regimen for more than 6 months with two consecu-
tive viral load determinations <75 copies/mL. Pa-
tients not receiving their first HAART treatment
SWITCH TO LPV/R MONOTHERAPY •PIERONE ET AL.239
regimen could be enrolled if prior regimens were
successful and if (a) changes made to prior regi-
mens were due only to lack of tolerability or to
achieve simplification or (b) the prior regimen had
been interrupted for any reason other than VF but
then restarted.
Patients were ineligible if any of the following
conditions were present: pregnancy or nursing;
concomitant therapy with drugs contraindicated
for use with LPV/r; vaccination within 4 weeks of
screening; active AIDS-defining opportunistic in-
fection or condition; recurrent episodes of moder-
ate-to-severe diarrhea or vomiting lasting 4 days
within 12 weeks prior to dosing; clinically signifi-
cant laboratory findings obtained during screening
including serum creatinine >2.0 mg/dL, alanine
transaminase (ALT), aspartate transaminase (AST),
or alkaline phosphatase levels >5 x upper limit of
normal (ULN), total bilirubin >2 x ULN, hemoglo-
bin <10 g/dL for males, <9.0 g/dL for females,
platelets <75,000 cells/mm3, and absolute neutro-
phil count <750 cells/mm3.
Data Analysis
The primary efficacy variable was the percent-
age of participants with plasma HIV RNA <75 cop-
ies/mL at 48 weeks. Virologic failure was defined
as HIV RNA levels >400 copies/mL on two con-
secutive occasions at least 1 week apart. Due to the
small sample size of the study, the exact 95% confi-
dence interval (CI) for the proportion of partici-
pants with plasma HIV-1 RNA <75 copies/mL at
48 weeks was computed.
The secondary efficacy variables included: per-
centage of participants with HIV RNA levels <75
copies/mL at 24 weeks; percentage of participants
with HIV RNA level <400 copies/mL at 24 and 48
weeks; the mean change in CD4+ lymphocyte
count at week 48; the mean change in lipid levels at
24 and 48 weeks; the percentage of participants
who required addition of or an increase in LLAs at
24 and 48 weeks; and percentage of participants
who discontinued therapy due to adverse events.
RESULTS
Baseline Characteristics
Twenty patients were screened between July and
November 2003. One patient was a screen failure
due to exacerbation of a chronic psychiatric illness.
One patient withdrew consent prior to the baseline
visit.
Eighteen patients were enrolled (13 men and 5
women). Five patients were African American and
13 were Caucasian. The mean age was 46.8 years.
The median time on prior HAART regimens was
122 weeks (range, 21–257 weeks). The median
baseline CD4+ cell count was 272 cells/mm3
(range, 73–1174 cells/mm3), and median nadir CD4
count was 177 cells/mm3 (range, 5–519 cells/mm3).
Prior NNRTI-based therapy included efavirenz
(EFV) in 4 (22%) cases and nevirapine (NVP) in 14
(78%) cases.
Participant Disposition and Study Outcomes
Study outcomes are presented in Tables 1 and 2.
Twelve of 18 (66%) participants met the primary
study endpoint with HIV RNA <75 copies/mL at
week 48 (95% CI 41.0–86.7). One participant (Pa-
tient 18) met the definition of VF at week 8 during a
period of nonadherence. After adherence counsel-
ing, the patient demonstrated virologic
resuppression and maintained a VL of <75 copies/
mL from weeks 24–48. Of the 18 participants en-
rolled, 13 (72%) completed 48 weeks of LPV/r
monotherapy and 12 of these 13 (92%) participants
on treatment at week 48 had plasma HIV RNA <75
copies/mL (Figure 1, Table 1). Overall, 10 (55%) of
the 18 participants maintained continuous viral
suppression (<75 copies/mL) at all time points
throughout the 48-week study period.
Two participants (Patients 5 and 15) exhibited
breakthrough viremia not meeting the definition of
VF. Patient 5 demonstrated a one time viral eleva-
tion (blip) at week 40 and the VL returned to <75
copies/mL by week 48. Patient 15 demonstrated
intermittent low-level viremia between weeks 8
and 48 (Table 1).
Twelve (92%) of 13 participants who completed
the study demonstrated an increase in CD4+ cell
count at week 48 (Table 1). For participants com-
pleting the study, the absolute CD4+ lymphocyte
count was 371 cells/mm3 at baseline and 462 cells/
mm3 at week 48, a mean increase of 91 cells/mm3
(Table 1).
Two (11%) participants (Patients 10 and 11) dem-
onstrated VF and were withdrawn from the study
(Table 2). Patient 10 demonstrated VF at week 32,
was withdrawn at that time point, and subse-
240 HIV CLINICAL TRIALS 7/5 SEPT-OCT 2006
quently was lost to follow-up. Patient 11 demon-
strated VF at week 16; was changed back to the
original regimen of tenofovir, lamivudine, and
nevirapine; attained viral resuppression after 4
weeks; and has maintained long-term control of
viremia. Three (17%) participants (Patients 7, 8,
and 13) withdrew early (weeks 2, 4, and 8, respec-
tively) due to diarrhea, all of whom had HIV RNA
plasma levels <75 copies/mL at the time of with-
drawal.
Viral Resistance Results
Of the three participants who demonstrated VF,
genotypic amplification was successful in two (Pa-
tients 10 and 11) (Table 3). Patient 10 had VF at
week 32, and genotypic mutations L63P and V77I
were identified. The phenotypic fold change was
0.57 to LPV. LPV trough concentrations from fro-
zen plasma specimens at weeks 2, 24, and 32 dem-
onstrated levels of 0.67 µg/mL, 1.95 µg/mL, and
3.26 µg/mL respectively (therapeutic range, 3–10
µg/mL). Patient 11 was noted to have an M36I
mutation at week 16, 3 weeks after VF. The LPV
trough concentration for Patient 11 at week 16 was
6.07 µg/mL. Patient 18 experienced VF at week 8
during a period of low adherence and demon-
strated virologic resuppression at the subsequent
visits. Testing for viral resistance was inadvert-
ently omitted at the time of VF. An LPV trough
concentration from a frozen plasma specimen col-
lected at week 24 was 8.27 µg/mL.
Adherence Monitoring Results
Pill counts demonstrated that 9 (69%) of the 13
participants completing the study maintained at
least an 80% level of adherence throughout 48
weeks (Table 1). Three of the four participants (Pa-
tients 5, 15, and 18) who maintained less than 80%
adherence experienced viral breakthrough. The
two participants (Patients 10 and 11) who were
prematurely withdrawn due to VF had greater
than 85% adherence to LPV/r throughout their
participation in the study (Table 2).
Adverse Events and Laboratory Abnormalities
Fourteen (78%) of the 18 participants reported
diarrhea, and three participants (17%) withdrew
due to mild or moderate diarrhea (Table 2). Four
participants (22%) experienced transient mild diar-
rhea that began within the first 2 weeks of taking
LPV/r and resolved by week 6. Seven participants
(39%) continued to have persistent mild diarrhea
over the course of the study.
Two (11%) participants (Patients 9 and 16) with
elevated blood glucose determinations in the 110–
125 mg/dL range at study entry developed an in-
crease in fasting glucose readings to over 125 mg/
dL and were diagnosed with diabetes mellitus. Pa-
tient 9 had elevated blood glucose determinations
throughout the study, but the hemoglobin A1C
level was within normal limits at week 12 and
medication was not deemed necessary. After study
completion, Patient 9 was sequentially changed to
an atazanavir/ritonavir-based HAART regimen
followed by the original regimen of Combivir
(lamivudine/zidovudine) and nevirapine. Despite
these changes in therapy, there was continued el-
evation of serum glucose and diabetic medication
Figure 1. Disposition of the 18 participants enrolled
who were switched from nonnucleoside reverse tran-
scriptase inhibitor/nucleoside reverse transcriptase in-
hibitor (NNRTI/NRTI) regimens to lopinavir/ritonavir
(LPV/r) monotherapy and were followed for 48 weeks.
DC = discontinued; VF = virologic failure.
18
patients
enrolled
3 patients 13 patients 2 patients
D/C completed D/C with VF
diarrhea 48 weeks
1 patient with
VF at week 8
weeks 24-48
1 patient
12/13 with
HIV RNA <75 HIV RNA
at week 48 1047 copies
at week 48,
no VF
SWITCH TO LPV/R MONOTHERAPY •PIERONE ET AL.241
Table 1. Study results of participants switched to LPV/r monotherapy who completed 48 weeks
HIV RNA levels, CD4 change,
Patient no. copies/mL Adherence, % cells/mm3
01 <75 for entire 48 weeks 96–100 274
02 <75 for entire 48 weeks 80–102 52
03 <75 for entire 48 weeks 96–101 30
04 <75 for entire 48 weeks 85–113 99
05 3314 at week 40; 61 at week 40; 7
otherwise <75 74–101 at other time
points
06 <75 for entire 48 weeks 83–100 50
09 <75 for entire 48 weeks 86–101 16
12 <75 for entire 48 weeks 81–101 114
14 <75 for entire 48 weeks 70–107 76
15 Intermittent viremia 37–100 52
16 <75 for entire 48 weeks 95–102 94
17 <75 for entire 48 weeks 83–102 189
18 10,068 at week 8 (VF); 81 and 58 at weeks 4 and 8; 233
<75 at weeks 24–48 59–76 at weeks 24–48
Note: LPV/r = lopinavir/ritonavir; VF = virologic failure.
was started. Patient 16 was started on diabetic
medication that led to blood glucose control, and
the patient completed 48 weeks of therapy. After
study completion, Patient 16 was changed back to
NNRTI-based therapy, glucose readings returned
to baseline, and antiglycemic medication was dis-
continued.
The National Cholesterol Education Program
(NCEP) guidelines were used for management of
lipid levels. Six (33%) of the 18 participants en-
rolled were receiving LLAs at study entry. Of the
13 participants who completed the trial, 7 (54%)
required the addition of or an increase in LLAs.
The mean cholesterol measurements in fasting
plasma samples for those seven participants were
225 mg/dL at baseline and 219 mg/dL at week 48
(change -6 mg/dL from baseline). The mean fast-
ing triglyceride measurements were 203 mg/dL at
baseline and 249 mg/dL at week 48 (change +46
mg/dL from baseline).
Six (46%) of the 13 participants completing the
trial had no change in LLAs. The mean fasting
cholesterol determinations were 183 mg/dL at
baseline and week 48 (+0 mg/dL). The mean fast-
ing triglyceride determinations at baseline were
136 mg/dL and 157 mg/dL at week 48 (change +21
mg/dL from baseline). None of the 13 participants
completing the trial experienced a grade 3 or 4
cholesterol or triglyceride elevation.
DISCUSSION
The appeal of PI monotherapy stems from a de-
sire to find a simple approach to HIV management
242 HIV CLINICAL TRIALS 7/5 SEPT-OCT 2006
that might avoid cumulative toxicities associated
with other antiretroviral classes. PI monotherapy
also has the potential to reduce the cost of long-
term therapy in certain settings. To that end, other
investigators have also conducted pilot studies to
evaluate the safety and efficacy of LPV/r
monotherapy. One study examined patients who
were receiving HAART but were naïve to PIs.19
Others looked at patients who were receiving
LPV/r-containing HAART20,21 and were simplified
to monotherapy. Yet another enrolled patients who
were naïve to HIV treatment.22 Finally, another
Table 2. Study outcomes of participants switched to LPV/r who did not complete 48 weeks
Patient Study HIV RNA levels, Adherence, CD4 change,
no. outcome copies/mL % cells/mm3
07 DC at week 4, <75 at week 4 100 14 at week 4
moderate diarrhea
08 DC at week 2 <75 at week 2 70 26 at week 2,
moderate diarrhea
10 VF at week 32, 6156 at week 28; 89–100 through 36 at week 32
LTFU 18,734 at week 32 week 28
11 VF at week 16, 1067 at week 12; 90–100 13 at week 12
resuppressed on 2278 at week 16;
previous regimen DC at week 16
13 DC at week 8, <75 at week 8 100 through week 4 151 at week 8
mild diarrhea
Note: LPV/r = lopinavir/ritonavir; DC = discontinued; VF = virologic failure; LTFU = lost to follow-up.
Table 3. Viral resistance results in participants with virologic failure (VF)
Weeks on VF LPV trough
Patient Week therapy with VF phenotype concentration,
no. of VF ongoing viremia genotype (LPV FC) µg/mL Study status
10 32 8 L63P, V77I 0.57 0.67 at week 2, LTFU at
1.95 at week 24, week 32
3.26 at week 32
11 12 3 M36I 0.8 (virtual) 6.07 at week 16 DC at
week 16
18 8 4 NA NA 8.27 at week 24 Resuppressed
Note: LPV FC = lopinavir fold change; LTFU = lost to follow-up; DC = discontinued; NA = not available.
study explored HAART-experienced patients with
multiple toxicities and treatment failures who were
placed on LPV/r monotherapy.23 Although the
numbers of patients studied in these initial pilot
trials were small, ranging from 6 to 21, the results
were encouraging; over 60% of participants by in-
tention-to-treat (ITT) analysis maintained viral
suppression from periods ranging from 24 to over
48 weeks. In these pilot trials, PI resistance was
rarely observed and few participants withdrew
due to treatment-related side effects.19–24
More recently, results from larger, randomized
SWITCH TO LPV/R MONOTHERAPY •PIERONE ET AL.243
comparative studies have been reported. These trials
include over 500 participants with follow-up ranging
from 48 to 96 weeks. Efficacy results again are similar
to the findings reported herein and the findings of the
aforementioned other small pilot trials (73%–89%
with HIV RNA levels <500 copies/mL).25–28
The results obtained from the current pilot study
of LPV/r monotherapy are encouraging and are
consistent with results reported by other investiga-
tors. Most of the participants maintained virologic
suppression and experienced an increase in CD4+
lymphocyte cell counts, and few withdrew due to
medication-related side effects.21,23,25,28 In the cur-
rent study, however, after switch to LPV/r, mild
diarrhea was commonly observed, increased blood
glucose occurred in two patients, and about half of
the participants needed the addition of or an in-
crease in lipid-lowering therapy. The costs for
management of these potential adverse effects
would need to be considered in any formal
pharmaco-economic analysis of LPV/r
monotherapy.
It is well known that resistance to LPV/r is very
rarely selected in PI-naïve patients who are started
on this agent,13,14,16,29,30 so it is not surprising that PI
resistance did not emerge during the study. The
mutations detected in the two patients with VF in
the current study have not been identified by the
International AIDS Society-USA as primary resis-
tance mutations for LPV.31 However, there have
been reports of LPV/r monotherapy leading to PI
genotypic resistance.24,32–35 The frequency of these
resistance cases among patients on LPV/r
monotherapy suggests that the barrier to PI resis-
tance may be lower with LPV/r monotherapy
when compared to LPV/r-based triple therapy.
Additionally, limited data suggest that mutations
in the Gag gene may influence resistance to some
PIs.36–38 The extent to which those findings may
relate to the VF of two participants (Patients 10 and
11) who maintained adherence of over 89% and
lacked evidence of significant genotypic or pheno-
typic resistance is unknown.
Another area of potential concern is the penetra-
tion of highly protein-bound LPV into the cere-
brospinal fluid (CSF).39 However, results of two
studies have shown that CSF levels of HIV RNA
decrease in patients receiving LPV/r as part of
HAART40 and LPV/r monotherapy.41 One study
using a highly sensitive assay with a lower limit of
detection of 3.7 µg/L level demonstrated that the
median CSF level of LPV/r in patients receiving
LPV/r-containing HAART exceeded the median
inhibitory concentration of HIV by 5-fold.42 Addi-
tional studies will be required to better understand
HIV central nervous system suppression in the
context of LPV/r monotherapy.
A comparison of costs between triple HAART
and LPV/r monotherapy reveals that LPV/r
monotherapy offers a potential cost savings in
some settings. The annual cost of one of the DHHS
recommended combination regimens purchased in
the United States1 for initial antiretroviral therapy
ranges from $12,323.88 to $15,935.88 compared
with $8,472 for LPV/r monotherapy. This repre-
sents a potential savings of between 31% and 47%
of drug acquisition costs,43 which is similar to the
findings of a pharmaco-economic analysis of LPV/r
monotherapy from Spain.44
This pilot trial was designed to provide prelimi-
nary data on the safety and efficacy of LPV/r
monotherapy rather than to support switching pa-
tients on successful NNRTI-based therapy to LPV/r
monotherapy. Our results suggest that LPV/r
monotherapy may offer a viable alternative for the
management of HIV infection. Future studies of
LPV/r monotherapy will utilize the LPV/r tablet
formulation (200 mg/50 mg) that reduces pill
count, does not require refrigeration, and may
have a lower incidence of diarrhea compared to the
soft gelatin capsule used in this trial.45 Results from
this pilot study and other trials reported to date
suggest that LPV/r monotherapy may be an option
for management of HIV infection. Larger, random-
ized trials will be required to define the patient
populations that might benefit most from LPV/r
monotherapy and to better understand the poten-
tial risks and benefits associated with this thera-
peutic strategy.
ACKNOWLEDGMENTS
Partial funding for this study was provided by
Abbott Laboratories, Abbott Park, Illinois, USA.
REFERENCES
1. Bartlett JG, Lane HC. Panel on Clinical Practices for Treat-
ment of HIV Infection, convened by the Department of
Health and Human Services (DHHS). Guidelines for the
Use of Antiretroviral Agents in HIV-Infected Adults and
Adolescents, October 2005. Available at: http://
244 HIV CLINICAL TRIALS 7/5 SEPT-OCT 2006
www.aidsinfo.nih.gov/guidelines/. Accessed November 15,
2005.
2. Palella FJ, Jr, Delaney KM, Moorman, AC, et al. Declining
morbidity and mortality among patients with advanced hu-
man immunodeficiency virus infection. N Engl J Med.
1998;338 (13):853–860.
3. Lewis W. Nucleoside reverse transcriptase inhibitors, mito-
chondrial DNA and AIDS therapy. Antivir Ther.
2005;10(suppl 2):M13–27.
4. Nolan D, Mallal S. Complications associated with NRTI
therapy: update on clinical features and possible pathogenic
mechanisms. Antivir Ther. 2004;9(6):849–863.
5. Galluzzi L, Pinti M, Guaraldi G, et al. Altered mitochondrial
RNA production in adiopocytes from HIV-infected individu-
als with lipodystrophy. Antivir Ther. 2005;10(suppl 2):M91–
99.
6. Antinori A, Zaccarelli M, Cingolani A, et al. Cross-resistance
among nonnucleoside reverse transcriptase inhibitors limits
recycling efavirenz after nevirapine failure. AIDS Res Hum
Retroviruses. 2002;18(12):835–838.
7. Bozzette SA, Joyce G, McCaffrey DF, et al. Expenditures for
the care of HIV-infected patients in the era of highly active
antiretroviral therapy. N Engl J Med. 2001;344(11):817–823.
8. Havlir DV, Marschner IC, Hirsch MS, et al. Maintenance
antiretroviral therapies in HIV infected patients with unde-
tectable plasma HIV RNA after triple-drug therapy. AIDS
Clinical Trials Group Study 343 Team. N Engl J Med.
1998;339(18):1261–1268.
9. Pialoux G, Raffi F, Brun-Vezinet F, et al. A randomized trial of
three maintenance regimens given after three months of
induction therapy with zidovudine, lamivudine and indinavir
in previously untreated HIV-1 infected patients. N Engl J
Med. 1998;339:1269–1276.
10. Reijers MH, Weverlling GJ, Jurriaans S, et al. Maintenance
therapy after quadruple induction therapy in HIV-1 infected
individuals: Amsterdam Duration of Antiretroviral Medication
(ADAM) study. Lancet. 1998;352:185–190.
11. Arieux C, Tattevin P, Souala FM, et al. Salvage therapy with
amprenavir and ritonavir: prospective study in 17 heavily
pretreated patients. HIV Clin Trials. 2002;3(2):125–132.
12. Duval X, Lamotte C, Race E, et al. Amprenavir inhibitory
quotient and virological response in human immunodefi-
ciency virus-infected patients on an amprenavir-containing
salvage regimen without or with ritonavir. Antimicrob Agents
Chemother. 2002;46(2):570–574.
13. Eron JJ, Feinberg J, Kessler HA, et al. Once-daily versus
twice-daily lopinavir/ritonavir in antiretroviral-naïve HIV-
positive patients: a 48-week randomized clinical trial. J In-
fect Dis. 2004;189(2):265–272.
14. Hicks C, King MS, Gulick RM, et al. Long-term safety and
durable antiretroviral activity of lopinavir/ritonavir in treat-
ment-naïve patients: 4 year follow-up study. AIDS.
2004;18(5):775–779.
15. Cvetkovic RS, Goa KL. Lopinavir/ritonavir: a review of its use
in the managment of HIV infection. Drugs. 2003;63(8):769–
802.
16. Perno CF, Cozzi-Lepri A, Balotta C, et al. Low prevalence of
primary mutations associated with drug resistance in antivi-
ral-naïve patients at therapy initiation. AIDS. 2002;16:619–
624.
17. Hsu A, Kempf D, Granneman R, et al. Exploring theoretical
mechanisms for lack of resistance to lopinavir/ritonavir in
antiretroviral naïve subjects [abstract 436-W]. In: Program
and abstracts of the 9th Conference on Retroviruses and
Opportunistic Infections, Seattle. Alexandria, VA: Founda-
tion for Retrovirology and Human Health; 2002:217.
18. Walsh JC, Mandalia, S, Gazzard, BG. Responses to a 1
month self-report on adherence to antiretroviral therapy are
consistent with electronic data and virological treatment
outcome. AIDS. 2002;16(2):269–277.
19. Ruane P, Luber A, Gaultier C, et al. Maintenance therapy
using lopinavir/ritonavir (LPV/r) alone for patients with well
controlled HIV-infection. In: Program and abstracts of the XV
International AIDS Conference; July 11–16, 2004; Bangkok,
Thailand. Abstract TuPeB4638
20. Campo RE, Lalanne R, Tanner TJ, et al. Lopinavir/ritonavir
maintenance monotherapy after successful viral suppres-
sion with standard highly active antiretroviral therapy in HIV-
1-infected patients. AIDS. 2005;19:447–452.
21. Arribas JR, Pulido F, Lorenzo A, et al. Lopinavir/ritonavir as
single-drug therapy for maintenance of HIV-1 viral suppres-
sion: 48-week results of a randomized, controlled, open-
label, proof-of-concept pilot clinical trial (OK Study). J
Acquir Immune Defic Syndr. 2005;40(3):280–287.
22. Gathe JC, Jr., Washington MY, Mayberry C, et al. IMANI-1
TC3WP single drug HAART-proof of concept study. Pilot
study of the safety and efficacy of Kaletra (LPV/r) as single
drug HAART in HIV + ARV-naïve patients-interim analysis of
subjects completing final 48 week data. In: Program and
abstracts of the XV International AIDS Conference; July 11–
16, 2004; Bangkok, Thailand. Abstract MoOrB1057.
23. Pierone G, Mieras J, Kantor C, et al. Lopinavir/r as sole
therapy for HIV-infection. In: Program and abstracts of the
9th European AIDS Conference; October 25–29, 2003; War-
saw, Poland. Abstract oral presentation Forum 1/5.
24. Pierone G, Mieras J, Kantor C, et al. Genotypic and pheno-
typic resistance observations among patients with viremia
while on lopinavir/ritonavir monotherapy. In: Program and
abstracts of the 44th Interscience Conference on Antimicro-
bial Agents and Chemotherapy; 2004; Washington, DC. Ab-
stract H-183.
25. Delfraissy J-F, Flandre P, Delaugerre C, et al. MONARK Trial
(MONotherapy AntiRetroviral Kaletra): 48-week analysis of
lopinavir/ritonavir (LPV/R) monotherapy compared to LPV/R
+ zidovudine/lamivudine (AZT/3TC) in antiretroviral-naïve
patients. In: Program and abstracts of the XVI International
AIDS Conference; August 13–18, 2006; Toronto, Canada.
26. Nunes WP, Oliveira MS, Almeida MMTB, et al. 48-week
efficacy and safety results of simplification to single agent
lopinavir/ritonavir (LPV/r) regimen in patients suppressed
below 80 copies/mL on HAART–the KalMo study in
antiretroviral-naïve patients. In: Program and abstracts of
the XVI International AIDS Conference; August 13–18, 2006;
Toronto, Canada.
27. Cameron W, da Silva B, Arribas J, et al. A two-year random-
ized controlled clinical trial in antiretroviral-naïve subjects
using lopinavir/ritonavir (LPV/r) monotherapy after initial in-
duction treatment compared to an efavirenz (EFV) 3-drug
regimen (Study MO3-613) in antiretroviral-naïve patients. In:
Program and abstracts of the XVI International AIDS Confer-
ence; August 13–18, 2006; Toronto, Canada.
28. Arribas J, Pulido F, Delgado R, et al. Lopinavir/ritonavir as
SWITCH TO LPV/R MONOTHERAPY •PIERONE ET AL.245
single-drug maintenance therapy in patients with HIV-1
suppression: forty-eight week results of a randomized,
controlled, open label, clinical trial (OKO4 Study) in
antiretroviral-naïve patients. In: Program and abstracts of
the XVI International AIDS Conference; August 13–18,
2006; Toronto, Canada.
29. Kempf D, King M, Bernstein B, et al. Incidence of resis-
tance in a double-blind study comparing lopinavir/ritonavir
plus stavudine and lamivudine to nelfinavir plus stavudine
and lamivudine. J Infect Dis. 2004;189:51–60.
30. Murphy R. Seven year follow-up of a lopinavir/ritonavir
(LPV/r)-based regimen in antiretroviral (ARV) naïve sub-
jects. In: Program and abstracts of the 10th European AIDS
Conference; November 17–20, 2005; Dublin, Ireland.
31. Johnson VA, Brun-Vezinet F, Clotet B, et al. Update of the
drug resistance mutations in HIV-1. Top HIV Med.
2006;14(3):125–130.
32. Friend J, Parkin N, Liegler T, et al. Isolated lopinavir resis-
tance after virological rebound of a ritonavir/lopinavir-
based regimen. AIDS. 2004;18:1965–1970.
33. Conradie F, Sanne I, Venter W, et al. Failure of lopinavir-
ritonavir (Kaletra)-containing regimen in an antiretroviral-
naïve patient. AIDS. 2004;18(7):1084–1085.
34. Goelz J, Wolf E, Moll A, et al. Single agent HAART with
lopinavir/r (LPV/r) in ART-naïve and pre-treated HIV-1-in-
fected patients. In: Program and abstracts of the XVI Inter-
national AIDS Conference; August 13–18, 2006; Toronto,
Canada.
35. Norton M, Delaugere C, Batot G, et al. Drug resistance
outcomes in a trial comparing lopinavir/ritonavir (LPV/r)
monotherapy to LPV/r + zivudine/lamivudine (MONARK
trial. In: Program and abstracts of the XV International HIV
Drug Resistance Workshop; June 13–17, 2006; Stiges,
Spain. Abstract 74.
36. Zhang YM, Imamichi H, Imamichi T, et al. Drug resistance
during indinavir therapy is casued by mutations in the
protease gene and its Gag substrate cleavage sites. J Virol.
1997;71(9):6662–6670.
37. Maguire MF, Guinea R, Griffin P, et al. Changes in human
immunodeficiency virus type 1 Gag at positions L449 and
P453 are linked to I50V protease mutants in vivo and cause
reduction of sensitivity to amprenavir and improved viral
fitness in vitro. J Virol. 2002;76(5):7398–7406.
38. Malet I, Roquebert B, Dalban C, et al. Association of Gag
cleavage sites to protease mutations and to virological
response in HIV-1 treated patients. J Infect. 2006; Jul 25.
Epub ahead of print.
39. Solas C, Lafeuillade A, Halfon P, et al. Discrepancies be-
tween protease inhibitor concentrations and viral load in
reservoirs and sanctuary sites in human immunodeficiency
virus-infected patients. Antimicrob Agents Chemother.
2003;47:238–243.
40. Lafeuillade A, Solas C, Halfon P, et al. Differences in the
detection of three HIV-1 protease inhibitors in non-blood
compartments: clinical correlations. HIV Clin Trials.
2002;3:27–35.
41. Van den Brande G, Beck JM, Capparelli E, et al. Kaletra
independently reduces HIV replication in cerebrospinal
fluid. In: Program and abstracts of the 12th Conference on
Retroviruses and Opportunististic Infections; February 22–
25, 2005; Boston, Massachusetts. Abstract 403.
42. Capparelli EV, Holland D, Okamoto C, et al. Lopinavir
concentrations in cerebrospinal fluid exceed the 50% in-
hibitory concentration for HIV. AIDS. 2005;19:949–952.
43. http://www.drugstore.com. Accessed January 15, 2006.
44. Escobar I, Pulido F, Arribas JR, et al. Pharmacoeconomic
analysis of lopinavir/ritonavir mono-therapy as mainte-
nance therapy in HIV-infected patients. In: Program and
abstracts of the 11th European AIDS Conference; Novem-
ber 17–20, 2005; Dublin, Ireland. Abstract PE19.5/2.
45. Zhu T, Chiu Y, Doan T, et al. New tablet formulation of
lopinavir/ritonavir is bioequivalent to the capsule at a dose
of 800/200 mg. In: Program and abstracts of the 45th
Interscience Conference on Antimicrobial Agents and Che-
motherapy (ICAAC); December 16–19, 2005; Washington,
DC.
... Although attempts dating from the late 1990s to decrease the number of drugs required to maintain HIV viral suppression have been unsuccessful (Havlir 1998;Reijers 1998), the use of boosted protease inhibitor (PI) single-drug monotherapy is now proving an effective new strategy to treat patients infected with HIV (Gathe 2004;Pulido 2008;Cameron 2008;Delfraissy 2008), decreasing the number of drugs used to treat HIV infection, thus reducing the cost of treatment (Escobar 2006) and hopefully minimizing the long-term toxicities of HAART (Cameron 2008). Recently published articles and reports suggest that PI-based monotherapy may be an effective therapeutic option for treatment of HIV infection (Ruane 2004;Campo 2005;Goelz 2006;Pierone 2006;Molto 2007;Arribas 2008). The efficacy and safety of PI-based monotherapy has been well documented and has shown promising results in controlled randomized clinical trials (RCTs), while economic studies in the U.K. and Spain have demonstrated advantages (Gazzard 2011; Arribas 2011). ...
Data
Full-text available
Despite the success of multiple-drug therapy regimens, the idea of treating human immunodeficiency virus (HIV) infection with fewer drugs is captivating due to issues of convenience, long-term toxicities and costs. This study investigated the impact on a local health budget of the introduction of a protease inhibitor (PI)-based antiretroviral monotherapy. An analysis of 23,721 administrative records of HIV-infected patients and a health technology assessment (HTA) were performed to assess cost-effectiveness, budget, organizational, ethics, and equity impact. Data showed that monotherapy had a annual cost of € 7,076 (patient with undetectable viral load) and € 7,860 (patient with detectable viral load), and that its implementation would realise economic savings of between 12 and 24 million euro (between 4.80% and 9.72% of the 2010 total regional budget expenditure for HIV management) in the first year, with cumulated savings of between 48 and 145 million euro over the following five years. Organizational, ethical and equity impact did not indicate any significant differences. The study suggests that for specific categories of patients monotherapy may be an alternative to existing therapies. Its implementation would not result in higher operating costs, and would lead to a reduction in total expenditure.
... Although attempts dating from the late 1990s to decrease the number of drugs required to maintain HIV viral suppression have been unsuccessful (Havlir 1998;Reijers 1998), the use of boosted protease inhibitor (PI) single-drug monotherapy is now proving an effective new strategy to treat patients infected with HIV (Gathe 2004;Pulido 2008;Cameron 2008;Delfraissy 2008), decreasing the number of drugs used to treat HIV infection, thus reducing the cost of treatment (Escobar 2006) and hopefully minimizing the long-term toxicities of HAART (Cameron 2008). Recently published articles and reports suggest that PI-based monotherapy may be an effective therapeutic option for treatment of HIV infection (Ruane 2004;Campo 2005;Goelz 2006;Pierone 2006;Molto 2007;Arribas 2008). The efficacy and safety of PI-based monotherapy has been well documented and has shown promising results in controlled randomized clinical trials (RCTs), while economic studies in the U.K. and Spain have demonstrated advantages (Gazzard 2011; Arribas 2011). ...
Article
Full-text available
Despite the success of multiple-drug therapy regimens, the idea of treating human immunodeficiency virus (HIV) infection with fewer drugs is captivating due to issues of convenience, long-term toxicities and costs. This study investigated the impact on a local health budget of the introduction of a protease inhibitor (PI)-based antiretroviral monotherapy. An analysis of 23,721 administrative records of HIV-infected patients and a health technology assessment (HTA) were performed to assess cost-effectiveness, budget, organizational, ethics, and equity impact. Data showed that monotherapy had a annual cost of € 7,076 (patient with undetectable viral load) and € 7,860 (patient with detectable viral load), and that its implementation would realise economic savings of between 12 and 24 million euro (between 4.80% and 9.72% of the 2010 total regional budget expenditure for HIV management) in the first year, with cumulated savings of between 48 and 145 million euro over the following five years. Organizational, ethical and equity impact did not indicate any significant differences. The study suggests that for specific categories of patients monotherapy may be an alternative to existing therapies. Its implementation would not result in higher operating costs, and would lead to a reduction in total expenditure.
... Modern combination antiretroviral therapy (cART) for HIVinfected drug naı¨venaı¨ve patients consists of a combination of three antiretroviral drugs from two classes, typically a boosted protease inhibitor or non-nucleoside analogue (NNTRI) in combination with two reverse transcriptase inhibitors (NRTI) [1]. Despite the remarkable success of dual class based cART [2] and the availability of multiple compound formulations allowing once daily intake with low pill burden [3], the concept of treating HIV-infected individuals with one single very potent drug for simplified maintenance therapy has attracted clinical HIV researchers over the past years [4,5]. The rational for this therapeutic approaches is the potential advantage of reduced adverse drug reactions, drugdrug interactions, reduced costs and the preservation of future treatment options in case of resistance related drug failure. ...
Article
Full-text available
The unparalleled success of combination antiretroviral therapy (cART) is based on the combination of three drugs from two classes. There is insufficient evidence whether simplification to ritonavir boosted protease inhibitor (PI/r) monotherapy in virologically suppressed HIV-infected patients is effective and safe to reduce cART side effects and costs. We systematically searched Medline, Embase, the Cochrane Library, conference proceedings and trial registries to identify all randomised controlled trials comparing PI/r monotherapy to cART in suppressed patients. We calculated in an intention to treat (loss-of follow-up, discontinuation of assigned drugs equals failure) and per-protocol analysis (exclusion of protocol violators following randomisation) and based on three different definitions for virological failure pooled risk ratios for remaining virologically suppressed. We identified 10 trials comparing 3 different PIs with cART based on a PI/r plus 2 reverse transcriptase inhibitors in 1189 patients. With the most conservative approach (viral load <50 copies/ml on two consecutive measurements), the risk ratios for viral suppression at 48 weeks of PI/r monotherapy compared to cART were in the ITT analysis 0.94 8 (95% CI 0.89 to 1.00) p = 0.06; risk difference -0.06 (95%CI -0.11 to 0) p = 0.05, p for heterogeneity  = 0.08, I(2) = 43.1%) and in the PP analysis 0.93 ((95%CI 0.90 to 0.97) p<0.001; risk difference -0.07 (95%CI -0.10 to -0.03) p<0.001, p for heterogeneity  = 0.44, I(2) = 0%). Reintroduction of cART in 44 patients with virological failure led in 93% to de-novo viral suppression. Virologically well suppressed HIV-infected patients have a lower chance to maintain viral suppression when switching from cART to PI/r monotherapy. Failing patients achieve high rates of de-novo viral suppression following reintroduction of reverse transcriptase inhibitors.
Article
The aim of the present study was to determine appropriate tenofovir-based regimens meriting evaluation in large-scale randomized trials among sub-Saharan African patients. Randomized, open-label, 96-week, prospective pilot study evaluating four first-line regimens: tenofovir/emtricitabine/nevirapine (Group 1), tenofovir/lopinavir/ritonavir (Group 2), tenofovir/emtricitabine/zidovudine (Group 3) and tenofovir/emtricitabine/efavirenz (Group 4) in antiretroviral-naïve, HIV-1-infected patients in Senegal and Cameroon. The primary endpoint was defined as an HIV-1-RNA viral load <50 copies/mL (study detection limit) at week 16 in at least 50% of patients using intention-to-treat analysis. At baseline, 119 patients included were 34% male, had a median plasma viral load of 5.4 log10 copies/mL and median CD4 cell count of 200/mm(3) (range=53-358). Primary endpoint was achieved for groups 1, 3, and 4 (58%, n=31; 62%, n=29; 53%, n=30, respectively), but not for Group 2 (38%, n=29). At week 96, undetectable HIV-1-RNA had been achieved in 74% of patients in Group 1, 38% Group 2, 72% Group 3 and 73% Group 4. Patients with detectable HIV-RNA at week 16 were more likely to have baseline HIV-1-RNA ≥100 000 copies/mL (adjusted OR=5.56; 95% CI:1.72-16.67). HIV-mutations associated with protease inhibitor resistance emerged in three patients, all of whom were in Group 2. Anemia occurred in two Group 3 patients and was the only serious treatment-related adverse event. Three efficient and safe tenofovir-based, triple regimens were identified; the two-drug regimen (tenofovir/lopinavir/r) did not achieve protocol-defined virologic threshold of efficacy.
Article
Background: Previous studies have evaluated the simplification of HIV treatment with ritonavir-boosted protease inhibitor monotherapy, demonstrating acceptable efficacy and advantages such as avoidance of the adverse effects of reverse transcriptase inhibitors. To achieve the best results, patients should be appropriately selected for this therapy. Objective: The purpose of this study was to estimate the proportion of HIV patients suitable for boosted protease inhibitor monotherapy according to clinical trial criteria. Setting The study was conducted in the outpatient hospital pharmacy service of the Complejo Hospitalario de Navarra in northern Spain. Method: A retrospective analysis was performed on data from 635 adults on antiretroviral therapy. The eligibility criteria were: (1) >18 years of age; (2) prior triple-drug antiretroviral regimen; (3) durability of current treatment >18 months; (4) viral load <400 copies/mL over the 18 months before evaluation and <50 copies/mL over the last 6 months; (5) CD4 count ≥250 cells/μL; (6) CD4 count nadir >100 cells/μL; (7) no previous virological failure under prior protease inhibitor-based regimen; (8) absence of co-infection with hepatitis B virus; (9) absence of HIV-related neurological disease; and (10) adherence >95 %. The average cost of the current treatment was calculated for patients who met all criteria, as well as the potential economic impact of simplification to monotherapy. Main outcome measure: Number of patients meeting all criteria for simplification to monotherapy according to clinical trial standards. Results: One hundred and three patients (16.5 %) met the clinical trial criteria for protease inhibitor monotherapy. One hundred and fifty patients (24 %) failed to fulfil only one of the conditions. Fifty-four percent of the patients who met all of the criteria had been treated for more than 10 years. The average saving per patient per year was 2,850- 3,400. Conclusion: This treatment strategy represents a realistic, albeit minority, option. Fulfilment of the above conditions should be the basis for simplification to protease inhibitor monotherapy, though the final decision depends on clinical criteria and patient preferences assessed by the attending physician. Further studies are needed to confirm long-term safety and efficacy.
Article
Treatment with lopinavir/ritonavir (LPV/r) monotherapy has been shown to be an effective alternative, especially in the maintenance of patients previously treated with combination therapy and prolonged virological suppression. LPV/r monotherapy is associated with a greater number of low-level viremia episodes than combination therapy, without resistance mutations being detected in the majority of patients. The incidence of the development of major resistance mutations in the OK pilot and OK04 studies was very low: 0.51 per 100 patients-year, and was mainly related to mutations in positions 46, 54 and 82, which have not compromised other therapeutic options. The contribution of low-level resistance mutations to loss of virological control seems small, and no different from that observed in combination therapy. However, this phenomenon should be studied in larger, long-term trials.
Article
Simplification of triple antiretroviral therapy to lopinavir/ritonavir (LPV/r) monotherapy in patients with well-controlled viremia for prolonged periods (more than 6 months) without prior failure with a protease inhibitor has been proposed as a strategy that could reduce the toxicity and costs of antiretroviral therapy in the long term while also preserving other therapeutic options. The results of several studies are currently available, some of which had a large number of patients and follow-up of up to 4 years.These studies indicate that this strategy is safe and efficacious, thus allowing its clinical use when indicated. This strategy may be especially useful in reducing the costs of treatment in countries with scarce economic resources. The role of LPV/r monotherapy in the prevention and management of lipodystrophy and in improving the selection of patients with an optimal riskbenefit ratio remains to be defined.
Article
Nine clinical trials have analyzed whether the use of lopinavir-ritonavir (LPV/r) monotherapy could be a valid alternative to triple antiretroviral therapy. Four of these clinical trials included an analysis of risk factors for virological failure. The MONARK study evaluated monotherapy in treatment-naïve patients. The M03-613 trial evaluated monotherapy after a period of induction therapy with triple combination antiretroviral therapy. The study by Sprinz et al and the OK studies evaluated monotherapy as maintenance of virological suppression. The efficacy of monotherapy varied according to the scenario. In the scenario of induction-maintenance, the factors related to virological failure were suboptimal adherence and low baseline CD4 counts. In the scenario of maintenance, the factors related to virological failure were suboptimal adherence, nadir CD4 count and low hemoglobin. In treatment-naïve patients, the risk of virological failure increased in patients who did not achieve a viral load of less than 400 copies/ml 4 weeks after initiating treatment and in those infected with non-B subtypes (a factor that was probably also related to suboptimal adherence).
Article
Despite the remarkable success of combination antiretroviral therapy, usually including the association of three antiretroviral drugs selected from two different classes, the possibility of treating HIV-infected patients with one single potent agent as simplified maintenance regimen has attracted clinicians and researchers over the past years. Monotherapy with one ritonavir-boosted protease inhibitor in subjects with persistently suppressed plasma HIV RNA offers several potential advantages, such as avoiding the long-term toxicity associated with nucleoside/nucleotide analogues, reducing costs, preventing drug-drug interactions, and preserving future treatment options. Several controlled and uncontrolled studies have assessed efficacy and safety of this monotherapy strategy, and the majority of available data concern lopinavir/ritonavir and darunavir/ritonavir. The virological efficacy of these boosted protease inhibitors as monotherapy is slightly lower than that of standard therapy, but the risk of resistance development is minimal and the re-introduction of nucleoside analogues usually leads to a re-suppression of the plasma viral load. Even though currently there is no consensus about the clinical use of protease inhibitor monotherapy for the treatment of HIV infection, this strategy seems an option for selected patients on stable combination therapy, with persistently suppressed plasma viral load, and without a history of virologic failure while receiving protease inhibitors. The aim of this article is to review and summarize the most recent randomized and observational studies which have evaluated efficacy and safety of the protease inhibitor monotherapy.
Article
Full-text available
Lopinavir (LPV) is an HIV protease inhibitor that is co-formulated with low-dose ritonavir (RTV), which enhances LPV pharmacokinetics (PK), and is marketed as Kaletra ® . • A dose of lopinavir/ritonavir (LPV/r) 800/200 mg once daily (QD) was recently approved in the United States for use in combination therapy for therapy-naïve HIV-infected adults. • LPV/r is currently available as 133.3/33.3 mg soft gelatin capsule (SGC) or 80/20 mg/mL liquid formulations, requires refrigerated storage prior to dispensing and is recommended to be taken with food in order to maximize lopinavir exposure. • A novel melt-extrusion technology was used to reduce pill count from 6 SGC per day to 4 tablets per day. In addition, this 200/50 mg tablet formulation of LPV/r does not require refrigeration. • Bioavailability and safety data of a single dose of LPV/r 400/100 mg administered as the tablet to healthy adults was previously reported. 1 – At a dose of 400/100 mg, the tablet provided LPV and RTV exposures similar to the SGC under fed (moderate fat meal) conditions. – The tablet also provided more consistent LPV and RTV exposures across meal conditions (fasting, moderate fat meal, high fat meal), with reduced pharmacokinetic variability compared to the SGC. Melt Extrusion Technology (Meltrex ™) LPV/r is a low solubility/low permeability drug (Biopharmaceutics Classification System Class 4). Historically, solid formulations of LPV/r showed poor bioavailability. • Unformulated solid fails to provide bioavailability (<5%). • Incorporation of surfactants, acids or other wetting agents with traditional technologies failed to provide adequate bioavailability for solid formulations. • In vitro dissolution did not necessarily correlate with in vivo bioavailability. Melt extrusion technology (Meltrex ™) has overcome these challenges. • Meltrex significantly improves the bioavailability of poorly soluble compounds like LPV/r by dissolving drug in polymer in a solvent-free environment. The drug remains in a state of molecular dispersion as the polymer hardens to form extruded material. • This extruded material can be further processed into conventional tablets. • Excipients used for the tablet are different than those in the SGC. Specifically, the tablet does not contain certain excipients (such as oleic acid, propylene glycol, sorbitol and castor oil) found in the SGC, which may contribute to gastrointestinal side effects. A single dose of 800/200 mg LPV/r administered as the tablet was bioequivalent, with respect to LPV, to 800/200 mg LPV/r as the SGC. The 90% confidence interval for LPV AUC and C max of the tablet compared to the SGC was within 0.80–1.25. The new LPV/r tablet formulation exhibited slightly higher bioavailability, approximately 17%, for both LPV and RTV following a single dose of 800/200 mg compared to the SGC.
Article
Full-text available
Background:Combination antiretroviral (ARV) therapy reduces HIV RNA levels in cerebro- spinal fluid (CSF) and the incidence of HIV-associated dementia (HAD). Despite this, the prevalence of HAD is rising, possibly because ARVs fail to completely suppress HIV in the brain. ARVs may fail because an important component of many regimens, protease inhibitors (PIs), extensively bind to plasma proteins, leaving little unbound drug to penetrate into the brain and CSF. However, these drugs are so potent that even low concentrations may inhibit HIV replication. The primary objective of this study was to determine whether a PI, Kaletra (LPV/r), reduces HIV RNA levels in CSF when used by itself. Methods:The study was an open-label, 24-week trial of sequential ARV therapy. Eighteen ARV-naive subjects underwent screening, which included measurement of HIV RNA in CSF and plasma and ARV resistance phenotype in plasma. Thirteen subjects enrolled and started on LPV/r alone for 3 weeks. At least 2 other ARVs were then added to complete 24 weeks. CSF was obtained again at weeks 3, 12, and 24. Data were analyzed using parametric and non-parametric tests, depending on data characteristics. CD4 counts are expressed in cells/µL and HIV RNA levels in log10 copies/mL. Results:At baseline, the median CD4 count was 225 and the median HIV RNA levels were 3.63 in CSF and 5.20 in plasma. Phenotype testing indicated the lack of resistance to all study drugs. Ten subjects completed the Week 3 visit, the primary study endpoint. After 3 weeks of LPV/r alone, HIV RNA declined in CSF (median 1.4, IQR 1.0-1.6, p
Article
Full-text available
The International AIDS Society-USA (IAS-USA) Drug Resistance Mutations Group reviews new data on HIV-1 drug resistance that have been published or presented at recent scientific meetings to maintain a current list of mutations associated with antiretroviral drug resistance.This December 2008 version of the IAS-USA drug resistance mutations figures updates those published in this journal in March/April 2008 (Johnson VA, Brun-Vezinet F, Clotet B, et al, Top HIV Med, 2008;16:62-68). The compilation includes mutations that may contribute to a reduced virologic response to HIV-1 drugs. It should not be assumed that the list presented here is exhaustive. Drugs that have been approved by the US Food and Drug Administration (US FDA) as well as any drugs available in expanded access programs are included and listed in alphabetical order by drug class. The figures are designed for practitioners to use in identifying key mutations associated with viral resistance to antiretroviral drugs and in making therapeutic decisions.
Article
Full-text available
Two different responses to the therapy were observed in a group of patients receiving the protease inhibitor indinavir. In one, suppression of virus replication occurred and has persisted for 90 weeks (bDNA, < 500 human immunodeficiency virus type 1 [HIV-1] RNA copies/ml). In the second group, a rebound in virus levels in plasma followed the initial sharp decline observed at the start of therapy. This was associated with the emergence of drug-resistant variants. Sequence analysis of the protease gene during the course of therapy revealed that in this second group there was a sequential acquisition of protease mutations at amino acids 46, 82, 54, 71, 89, and 90. In the six patients in this group, there was also an identical mutation in the gag p7/p1 gag protease cleavage site. In three of the patients, this change was seen as early as 6 to 10 weeks after the start of therapy. In one patient, a second mutation occurred at the gag p1/p6 cleavage site, but it appeared 18 weeks after the time of appearance of the p7/p1 mutation. Recombinant HIV-1 variants containing two or three mutations in the protease gene were constructed either with mutations at the p7/p1 cleavage site or with wild-type (WT) gag sequences. When recombinant HIV-1-containing protease mutations at 46 and 82 was grown in MT2 cells, there was a 68% reduction in its rate of replication compared to the WT virus. Introduction of an additional mutation at the gag p7/p1 protease cleavage site compensated for the partially defective protease gene. Similarly, rates of replication of viruses with mutations M46L/I, I54V, and V82A in protease were enhanced both in the presence and in the absence of Indinavir when combined with mutations in the gag p7/p1 and the gag p1/p6 cleavage sites. Optimal rates of virus replication require protease cleavage of precursor polyproteins. A mutation in the cleavage site that enhanced the availability of a protein that was rate limiting for virus maturation would confer on that virus a significant growth advantage and may explain the uniform emergence of viruses with alterations at the p7/p1 cleavage site. This is the first report of the emergence of mutations in the gag p7/p1 protease cleavage sites in patients receiving protease therapy and identifies this change as an important determinant of HIV-1 resistance to protease inhibitors in patient populations.
Article
The long-term effectiveness of potent three-drug antiretroviral regimens for the treatment of human immunodeficiency virus type 1 (HIV-1) infection is limited by problems related to compliance and tolerability. We investigated whether two-drug maintenance therapy would suppress viral replication after a three-month period of aggressive triple-drug induction therapy. A total of 378 HIV-1-infected adults who had not received previous antiretroviral treatment received three months of induction therapy consisting of 300 mg of zidovudine every 12 hours, 150 mg of lamivudine every 12 hours, and 800 mg of indinavir every 8 hours. The 279 patients in whom the plasma HIV-1 RNA titer fell below 500 copies per milliliter after two months of triple-drug therapy, and who completed the induction phase, were randomly assigned at month 3 to one of the following three open-label maintenance regimens: zidovudine, lamivudine, and indinavir; zidovudine and lamivudine; or zidovudine and indinavir. The primary end point was an increase in HIV-1 RNA levels to 500 copies or more per milliliter during the maintenance phase. The proportion of patients who reached the primary end point was significantly higher among patients receiving zidovudine plus lamivudine (29 of 93 patients, P<0.001) or zidovudine plus indinavir (21 of 94, P=0.01) than among patients receiving continued triple-drug therapy (8 of 92). This higher failure rate in the groups treated with the two-drug maintenance regimens was also observed in the subgroup of patients with maximally suppressed HIV-1 RNA (below 50 copies per milliliter) at the time of randomization to maintenance therapy. In HIV-1-infected adults not previously treated with antiretroviral drugs whose plasma HIV-1 RNA levels fell below 500 copies per milliliter after three months of induction therapy with zidovudine, lamivudine, and indinavir, two-drug maintenance therapy was less effective in sustaining a reduced viral load than continued three-drug therapy.
Article
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 per cubic millimeter, who were seen at nine clinics specializing in the treatment of human immunodeficiency virus (HIV) infection in eight U.S. cities from January 1994 through June 1997. Mortality among the patients declined from 29.4 per 100 person-years in the first quarter of 1995 to 8.8 per 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 three major opportunistic infections (Pneumocystis carinii pneumonia, Mycobacterium avium complex disease, and cytomegalovirus retinitis) declined from 21.9 per 100 person-years in 1994 to 3.7 per 100 person-years 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. The recent declines in morbidity and mortality due to AIDS are attributable to the use of more intensive antiretroviral therapies.
Article
Highly active antiretroviral therapy (HAART) has led to health benefits for patients infected with HIV-1. However, long-term use of multidrug regimens is difficult to sustain. Simplifying antiretroviral treatment regimens would increase patients' adherence and minimise toxicity. We investigated the feasibility of a strategy of induction therapy followed by maintenance therapy with HAART in a randomised open-label study. From March, 1997, we enrolled patients infected with HIV-1 with at least 200 CD4 cells/microL, at least 1000 HIV-1 RNA copies/mL in plasma, and no previous exposure to antiretroviral drugs. After 26 weeks of induction therapy (stavudine, lamivudine, saquinavir, and nelfinavir) patients were randomly allocated maintenance therapy (either stavudine and nelfinavir or saquinivir and nelfinavir) or prolonged induction therapy (if the plasma HIV-1 RNA concentration at weeks 24 and 25 was <50 copies/mL). In February, 1998, we discontinued randomisation after an interim analysis. 62 patients had been enrolled. 39 (91%) of the 43 patients who were followed up for at least 26 weeks had an undetectable plasma HIV-1 RNA concentration at week 16. At week 26, 31 patients were randomly allocated treatment. Of these patients 25 had a total follow-up of at least 36 weeks. At week 36, a higher proportion of patients on maintenance therapy (nine [64%] of 14) had a detectable HIV-1 RNA than patients on prolonged induction therapy (one [9%] of 11, p=0.01). The initial virion-clearance rate during induction therapy was higher in five patients on maintenance therapy with a sustained undetectable plasma HIV-1 RNA concentration than in nine patients with recurrence of a detectable plasma HIV-1 RNA concentration at week 36 (0.35 vs 0.19 per day, respectively; p=0.0008). The induction regimen provided a rapid suppression of viral replication to below 50 copies/mL. However, suppression was not sustained in a considerable number of patients who went onto maintenance therapy. It is currently inadvisable to continue attempts at moving from induction to maintenance therapy in day-to-day practice.
Article
Combination antiretroviral therapy with indinavir, zidovudine, and lamivudine can suppress the level of human immunodeficiency virus (HIV) RNA in plasma below the threshold of detection for two years or more. We investigated whether a less intensive maintenance regimen could sustain viral suppression after an initial response to combination therapy. HIV-infected subjects who had CD4 cell counts greater than 200 per cubic millimeter, who had been treated with indinavir, lamivudine, and zidovudine, and who had less than 200 copies of HIV RNA per milliliter of plasma after 16, 20, and 24 weeks of induction therapy were randomly assigned to receive either continued triple-drug therapy (106 subjects), indinavir alone (103 subjects), or a combination of zidovudine and lamivudine (107 subjects). The primary end point was loss of viral suppression, which was defined as a plasma level of at least 200 copies of HIV RNA per milliliter on two consecutive measurements during maintenance therapy. During maintenance treatment, 23 percent of the subjects receiving indinavir and 23 percent of those receiving zidovudine and lamivudine, but only 4 percent of those receiving all three drugs, had loss of viral suppression (P<0.001 for the comparison between triple-drug therapy and the other two maintenance regimens). Subjects with greater increases in CD4 cell counts during induction therapy, higher viral loads at base line (i.e., at the beginning of induction therapy), and slower rates of viral clearance were at greater risk for loss of viral suppression. The presence of zidovudine-resistance mutations in HIV RNA at base line was strongly predictive of the loss of viral suppression in subjects treated with zidovudine and lamivudine. The suppression of plasma HIV RNA after six months of treatment with indinavir, zidovudine, and lamivudine is better sustained by the continuation of these three drugs than by maintenance therapy with either indinavir alone or zidovudine and lamivudine.