ArticlePDF Available

New Options in the Treatment of Lipid Disorders in HIV-Infected Patients

Authors:

Abstract and Figures

Since the introduction of HAART, there was a remarkably change in the natural history of HIV disease, leading to a notable extension of life expectancy, although prolonged metabolic imbalances could significantly act on the longterm prognosis and outcome of HIV-infected persons, and there is an increasing concern about the cardiovascular risk in this population. Current recommendations suggest that HIV-infected perons undergo evaluation and treatment on the basis of the Third National Cholesterol Education Program Expert Panel on Detection, Evaluation and Treatment of High Blood Cholesterol in Adults (NCEP ATP III) guidelines for dyslipidemia, with particular attention to potential drug interactions with antiretroviral agents and maintenance of virologic control of HIV infection. While a hypolipidemic diet and physical activity may certainly improve dyslipidemia, pharmacological treatment becomes indispensable when serum lipid are excessively high for a long time or the patient has a high cardiovascular risk, since the suspension or change of an effective antiretroviral therapy is not recommended. Moreover, the choice of a hypolipidemic drug is often a reason of concern, since expected drug-drug interactions (especially with antiretroviral agents), toxicity, intolerance, effects on concurrent HIV-related disease and decrease patient adherence to multiple pharmacological regimens must be carefully evaluated. Often the lipid goals of patients in this group are not achieved by the therapy recommended in the current lipid guidelines and in this article we describe other possibilities to treat lipid disorders in HIV-infected persons, like rosuvastatin, ezetimibe and fish oil.
Content may be subject to copyright.
The Open AIDS Journal, 2009, 3, 31-37 31
1874-6136/09 2009 Bentham Open
Open Access
New Options in the Treatment of Lipid Disorders in HIV-Infected Patients
Erika Ferrari Rafael da Silva*,1 and Giuseppe Bárbaro2
1Federal University of São Paulo, R Loefgren, 1588, Zip Code04040 002, São Paulo, Brazil
2Cardiology Unit, Department of Medical Pathophysiology, University La sapienza, Rome, Italy
Abstract: Since the introduction of HAART, there was a remarkably change in the natural history of HIV disease, leading
to a notable extension of life expectancy, although prolonged metabolic imbalances could significantly act on the long-
term prognosis and outcome of HIV-infected persons, and there is an increasing concern about the cardiovascular risk in
this population. Current recommendations suggest that HIV-infected perons undergo evaluation and treatment on the basis
of the Third National Cholesterol Education Program Expert Panel on Detection, Evaluation and Treatment of High Blood
Cholesterol in Adults (NCEP ATP III) guidelines for dyslipidemia, with particular attention to potential drug interactions
with antiretroviral agents and maintenance of virologic control of HIV infection. While a hypolipidemic diet and physical
activity may certainly improve dyslipidemia, pharmacological treatment becomes indispensable when serum lipid are
excessively high for a long time or the patient has a high cardiovascular risk, since the suspension or change of an
effective antiretroviral therapy is not recommended. Moreover, the choice of a hypolipidemic drug is often a reason of
concern, since expected drug-drug interactions (especially with antiretroviral agents), toxicity, intolerance, effects on
concurrent HIV-related disease and decrease patient adherence to multiple pharmacological regimen s must be carefully
evaluated. Often the lipid goals of patients in this group are not achieved by the therapy recommended in the current lipid
guidelin es and in this article we describe other possibilities to treat lipid disorders in HIV-in fected persons, like
rosuvastatin, ezetimibe and fish oil.
Keywords: Human immunodefciency virus, acquired immunodeficiency syndrome, highly active antiretroviral therapy,
dyslipidemia, statins, fish oil, ezetimibe.
INTRODUCTION
Disorders of lipid metabolism have been described in
patients with HIV infection before the introduction of highly
active antiretroviral therapy (HAART), includ ing increased
serum triglyceride (TG) levels and decreased cholesterol
levels observed in advanced stages of HIV infection [1-3].
However, laboratory and clinical abnormalities of lipid
metabolism have also been increasingly recognized after the
advent of HAART. Significant increases in plasma TG and
total cholesterol (TC) concentrations, often associated with
abnormal body fat distribution and glucose metabolism
alterations (such as peripheral insulin resistance,
hyperinsulinemia, hyperglycemia and diabetes mellitus),
have been reported specially in protease inhibitors (PI)-
treated patients [4-6]. For details see Table 1. Since the
introduction of HAART, there was a remarkably change in
the natural history of HIV disease, leading to a notable
extension of life expectancy, although prolonged metabolic
imbalances could significantly act on the long-term
prognosis and outcome of HIV-infected persons, and there is
an increasing concern about the cardiovascular risk in this
population [4,7,8]. While a hypolipidemic diet and physical
activity may certainly improve dyslipidemia,
pharmacological treatment becomes indispensable when
*Address correspondence to this author at the Federal University of São
Paulo, R Loefgren, 1588, Zip Code04040 002, São Paulo, Brazil; Tel: 55 11
5081 8972; Fax: 55 11 5081 8972;
E-mails: erikaferrari @uol.com.br, ferrarierika76@hotmail.com
serum lipid are excessively high for a long time or the
patient has a high cardiovascular risk, since the suspension
or change of an effective antiretroviral therapy is not
recommended [9]. Moreover, the choice of a hypolipidemic
drug is often a reason of concern, since expected drug-drug
interactions (especially with antiretroviral agents), toxicity,
intolerance, effects on concurrent HIV-related disease and
decrease patient adherence to multiple pharmacological
regimens must be carefully ev aluated [8,10]. HIV infected
person have an increase risk of coronary artery d isease
(CAD) [11]. Part of this risk may be due to the
hyperlipidemia associated with antiretroviral treatment
[11,12]. Often the lipid goals of patients in this group are not
achieved by the therapy recommended in the current lipid
guidelines [3] and in this article we describe others
possibilities to treat lipid disorders in HIV-infected persons.
PATHOGENESIS FOR DYSLIPIDEMIA
PIs- Associated Metabolic Alterations
PIs target the catalytic region of HIV-1 protease. This
region is homologous with regions of two human proteins
that regulate lipid metabolism: cytoplasmic retinoic-acid
binding protein-1 (CRABP-1) and low density lipoprotein-
receptor-related protein (LRP) [13,14]. It is hypothesized
that PIs inhibit CRABP-1-modified and cytrochrome P450
3- mediated synthesis of cis-9 retinoic acid and peroxisome
proliferator activated receptor (PPAR ) heterodimer. The
inhibition increases the rate of apoptosis of adipocytes and
reduces the rate at which pre-adipocytes differentiate into
32 The Open AIDS Journal, 2009, Volume 3 da Silva and Bárbaro
adipocytes, with the final effect of reducing TG storage and
increasing lipid release. PIs-binding to LRP would impair
hepatic chylomicron uptake and endothelial TG clearance,
resulting in hyperlipidemia and insulin resistance [13,14].
Table 1. Changes in Lipid Metabolism in HIV Infection
HIV-Infected Naïve Patients
Triglyceride
VLDL
VLDL triglyceride production rates
Small, dense LDL
HIV Patients Treated with IP Based Regimen
Triglyceride
Total Cholesterol
VLDL, IDL, LDL-c, cholesterol Postprandial delipidation
Apo B-100 VLDL to IDL/LDL transfer
Apo E VLDL and LDL catabolic rates
ApoC-III Hepatic lipase activity
VLDL production Lipoprotein lipase activity
VLDL - very low-density lipoprotein (LDL), HDL – high density lipoprotein, IDL –
intermediate-density lipoprotein, Apo – apolipoprotein.
Some data indicate that PI-associated dyslipidemia may
be caused, at least in part, by PI-mediated inhibition of
proteasome activity and accumulation of the active portion
of sterol regulatory element-binding protein (SREBP)-1c in
liver cells and adipocytes or by apoliprotein (apo) CIII
polymorphisms in HIV-infected persons [15]. The observed
excess of apo CIII in lipoprotein might be a major
determin ant of a slower catabolism of triglyceride-rich
lipoproteins because apo CIII is an inhibitor of lipoprotein
lipase activity and also impairs the interaction of apo B and
apo E with LDL receptor and LRP. This will result in an
increased level of remnant lipoprotein returning to the liver
[15,16]. Sequence homologies have been described between
HIV-1 protease and human site-1 protease (S1P), which
activates SREBP-1c and SREBP-2 pathways. A
polymorphism in the S1P/SREBP-1c genes confers a
difference in risk for development of an increase in TC with
PI therapy, suggesting a possible genetic predisposition to
hyperlipoproteinemia in PI-treated patients [17]. Caron et al.
[17] reported that some PIs may impair the nuclear location
of SREBP-1 and alter the structure and stability of the
nuclear lamina possibly by impairing the maturation of
prelamin A to lamin A. Lamin A and lamin C are encoded
by the same gene and can combine with lamin B to form a
network of filamentous proteins located at the inner face of
the nuclear membrane called lamina [17]. Lamina interacts
with the nuclear membrane and with chromatin. In
particular, the C-terminal globular domain of lamin A/C can
bind DNA and also SERBP-1 [17]. Cells from these patients
have nuclear alterations and altered lamina stability, similar
to the alterations induced in cultured adipocytes by some PIs
[17]. It is possible to hypothesize that some PIs may alter the
lamina structure and thereby alter the normal location of
SREBP-1 inside the nucleus [17]. This could impair
adipocyte differentiation and induce insulin resistance.
Moreover, some PIs may alter the expression of
adipocytokines in cultured adipocytes [18], increasing the
expression of proinflammatory cytokines TNF- and IL-6,
which are known to play a pathogenetic role in adipose
tissue apoptosis and decreasing the expression of
adiponectin, which is associated with the development of
insulin resistance [18].
Treatment Considerations
The studies published until now show that dyslipidemia
in HIV-infected persons carries the same degree of
cardiovascular risk as in HIV-negative population [19].
Nowadays the benefits of lipid-lowering interventions have
been extended to HIV-infected persons. Further, there is
currently no basis for a more aggressive intervention among
HIV-infected persons than what is currently recommended
for the general population. Because there is a significant
possibility for drug interaction of some lipid-lowering agents
with antiretroviral drugs (in particular statins are metabolized
through the cytochrome (CYP) P450 system), care should be
given to the choice of lipid-lowering agents. Current
recommendations suggest that HIV-infected persons undergo
evaluation and treatment on the basis of the Third National
Cholesterol Education Program Expert Panel on Detection,
Evaluation and Treatment of High Blood Cholesterol in
Adults (NCEP ATP III) guidelines for dyslipidemia, with
particular attention to potential drug interactions with
antiretroviral agents and maintenance of virologic control of
HIV infection. Nonpharmacologic measures remain the basis
for intervention. The initial choice for hypertriglyceridemia
is fibrate, and for elevated LDL-c, statins [20].
According to the US-based Adult AIDS Clinical Trial
Group (AACTG) Cardiovascular Disease Focus Group,
treatment with pravastatin, fluvastatin or atorvastatin is
recommended for antiretroviral-linked hypercholesterolemia,
while lovastatin and simvastatin should be avoided due to
interactions with PIs or non-nucleoside reverse transcriptase
inhibitors (NNRTI) [21] and the risk of skeletal muscle
toxicity. Many studies that evaluate the effect of statins for
the treatment of antiretroviral-associated dyslipidemia have
shown only partial responses to such therapy, with total and
LDL-c values being reduced by just 25% [22].
Statins are considered the current first-line therapy for
primary hypercholesterolemia and showed beneficial effects
in both reducing total and LDL-c cholesterol levels in the
HIV-negative persons [23]. Because of the variable
metabolism through CYP 3A4, significant interactions have
been documented with potent CYP 3A4 inhibitors (such as
itraconazole, cyclosporine, oral anticoagulants, PIs and
delaverdine), which cause elevated levels of statins, leading
to a significantly increase of liver and skeletal muscle
toxicity [23]. Lovastatin and simvasatin are administered as
inactive lactone prodrugs that are avidly metabolized by
intestinal and hepatic CYP 3A4. On the other hand,
pravastatin, atorvastatin and fluvastatin are administered
directly as the active hydroxyl-acid. Pravastatin is eliminated
mostly by glucoronidation, fluvastatin by CYP 2C9 isoform,
and CYP 3A4 has no role in their metabolism [24]. On the
basis on the clinical and experimental data, simvastin and
lovastatin should not be used in patients taking PIs or
Treatment of Lipid Disorders in HIV-Infected People The Open AIDS Journal, 2009, Volume 3 33
NNRTIs, atorvastatin can be used with caution (at low initial
doses) and pravastatin and fluvastatin appear to be safe for
use in association with HAART [19]. Table 2 has a summary
of the lipid lowering therapy in HIV infected persons.
Rosuvastatin
Rosuvastatin is a 3-hydroxy-3-methylglutaryl coenzyme
A (HMG-CoA) inhibitor that showed the highest dose-to-
dose potency in lowering total and LDL-c levels, compared
with other currently available statins. It works also to reduce
TG and increase HDL-c. Moreover, pharmacokinetic studies
have demonstrated that its metabolism is not dependent on
the CYP 450 3A4 isoenzyme and its use could be considered
in PI-treated individuals as the result of the low risk of drug-
drug interactions [25]. Only 10% of the administered dose is
metabolized by CYP 2C9 isoenzyme into N-desmethyl
rosuvastatin and its metabolite are 90% eliminated by the
fecal route [24-26]. The usual recommended starting dose of
rosuvastatin is 10 mg daily, but initiation at 5 mg daily may
be considered for patients who have predisposing factors for
myopathy or are taking cysclosporine. In subjects with
severe renal impairment or taking fibrates, therapy with
rosuvastatin should only be used with great caution, daily
dose should be initiated at 5 mg and not exceed 10 mg [26].
In the research of Bottero et a l. [27], the use of
rosuvastatin (10 mg/day) was retrospective evaluated during
16 weeks in HAART-treated HIV-infected persons with
dyslipidemia. Seventy eight patients started on rosuvastatin,
sixty as monotherapy. After 16 weeks of treatment, a
significant decrease was seen in both LDL-c and non-HDL
(31,3% and 29,9% reduction respectively). The decrease in
triglyceride was also significant (34,1%). Rosuvastatin was
safe and effective in the treatment of dyslipidemia in
HAART-treated HIV-infected persons. The results showed
above are similar to the HIV-uninfected population. Calza et
al. [28] in an open label, randomized, prospective study
evaluated the role of rosuvastatin (10 mg once daily),
pravastatin (20 mg once daily) and atorvastatin (10 mg once
daily) in the treatment of hypercholesterolemia (TC > 250
mg/dL) in HIV-infected persons at least 3 month duration
and unresponsive to a hypolipidemic diet and physical
exercise. Eighty five subjects completed the study and the
mean reduction after one year follow up was 21,2% and
23,6% versus baseline TC and LDL-c levels, respectively
(p=0.002). Mean decrease in TC concentration was
significantly greater with rosuvastatin (25,2%) than with
pravastatin (17,6% p = 0.01) and atorvastatin (19,8% p =
0.03). All the statins showed a favourable tolerability profile,
but rosuvastatin was found to be more effective. Van der Lee
et al. [29] evaluated the pharmacokinectics and
pharmacodynamics of combined use of lopinavir/ritonavir
and rosuvastatin (10 mg, 20 mg and 40 mg) in HV-infected
persons and found that the levels of this PI were not affected
by the administration of rosuvastatin, but the rosuvastin
levels unexpectedly appeared to be 1.6 fold compared with
healthy volunteers. Larger studies are necessary to confirm
these findings, until there, the combination of rosuvastin and
lopinavir/ritonavir should be used with caution.
Ezetimibe
Ezetimibe is the first lipid-lowering drug that inhibits
intestinal uptake of dietary and biliary cholesterol at the
brush border of the intestine, resulting in a reduction of
hepatic cholesterol stores and an increase in clearance of
cholesterol form in the blood [30]. It doesn’t affect the
absorption of fat-soluble nutrients and is an attractiv e option
for HIV-infected persons because it lacks CYP P450
metabolism and therefore is not expected to interact with
antiretroviral [30,31]. It reduces cholesterol absorption in the
duodenum by approximately 50%, thereby attaining
reductions in LDL-c of 20% [32]. This benefit is
significantly greater when it is given with any of the statins,
achieving reductions in LDL-c of up to 50% [32,33]. This
synergistic effect of the two drugs in combination results
from the inhibition of duodenal cholesterol absorption by
ezetimibe, together with the reduction of hepatic cholesterol
production by the statins [30]. Following oral administration,
Table 2. Lipid Lowering Therapy for HIV-Infected People
Lipid Alteration Therapy Caution
Elevated LDL-c or
non-HDL
cholesterol with
triglycerides level of
200-500 mg/dL
Statins:
- Pravastatin: 20-40 mg daily
- Atorvastatin: 10-20 mg/daily
- Fluvastatin: 20-40 mg/daily
- Rosuvastatin
- Lovastatin
- Simvastatin
- Ezetimibe: 10 mg daily
Less drug interaction potential
Used with caution in lower doses when combined with PIs and NNRTIs
Minimal drug interaction potential. Not widely used due to low potency
Most potent statin. May be used safely with antiretroviral
Avoid in patients taking PIs
Avoid in patients taking PIs or delaverdine
Can be used with statin or in monotherapy
triglycerides level >
500 mg/dL
Fibrates and Fish Oil:
- Gemfibrozil: 1200 mg daily
- Fenofibrate: 200 mg daily
- Bezafibrate: 400 mg daily
- Omega-3 polyunsaturated fatty
acids/fish oil: 3-5g
Caution wh en used with statins in mixed dyslipid emia
Caution wh en used with statins in mixed dyslipid emia
First line therapy for hypertriglyceridemia
Evidence suggest fish oil may decrease triglycerides and increase HDL-c,
however can also increase LDL-c
Adapted from Bennet M [38], Stebbing J [39], Calza L [53], Soler A [54], Bader MS [55].
34 The Open AIDS Journal, 2009, Volume 3 da Silva and Bárbaro
ezetimibe is rapidly absorbed and extensively metabolized
(>80%) to the pharmacolog ically active ezetimibe-
glucuronide [30]. The recommended dose is 10 mg/day, and
can be administered in the morning or evening without
regard to food [30]. The major metabolic pathway for
Ezetimibe consists of glucuronidation of 4-hydroxyphenyl
group by uridine 5’-diphosphate-glucuronosyltransferase
isoenzymes to form ezetimibe-glucuronide in the intestine
and liver [30]. It has a favourable drug-drug interaction
profile. Ezetimibe does not have significant effects on
plasma levels of HMG-CoA reductase inhibitors known as
statins (atorvastatin, fluvastatin, lovastatin, pitavastatin,
pravastatin, rosuvastatin, simvastatin), fibric acid derivatives
(gemfibrozil, fenofibrate), digoxin, glipizide, warfarin and
triphasic oral contraceptives (ethinylestradiol an d
levonorgestrel). Concomitant administration of food,
antiacids, cimetidine or statins had no significant effect on
ezetimibe bioavailability [34].
Ezetimibe has been effective in optimizing lipid levels
when added to traditional therapy in non-HIV infected
persons [34-36]. In HIV positive population some studies
have assessed the efficacy of this drug and are described as
follow. Coll et al. [37] showed that ezetimibe monotherapy
decreases LDL-c as effectively as fluvastatin monotherapy.
Twenty HIV-infected persons were randomly assigned to
receive ezetimibe (10 mg/day) or fluvastatin (80 mg/day).
Patients receiving ezetimibe experienced a statistically
significant (p=0,003) 20% reduction in the concentration of
LDL-c, similar to that observed with fluvastatin (24%, p
between groups 0.70). Negredo et al. [30] conducted a
prospective study and Ezetimibe was added when the statin
(pravastatin monotherapy) has poor response. During 24
weeks, nineteen patients received ezetimibe (10mg/day) and
pravastatin (20 mg/day), while the patients maintained the
same antiretroviral regimen. At week 24, 61,5% of patients
achieved the endpoint of the study (LDL-c < 130 mg/dL).
Significant declines in total and LDL-c levels were observed
between baseline and weeks 6, 12, 24, irrespective of
antiretroviral regimen (IP or ITRNN) and mean HDL-c
increased significantly. No patients discontinued therapy due
to intolerance or toxicity. The addition of ezetimibe to
ongoing pravastin was effective and a safe option for HIV-
infected persons not achieving the NCEP ATP III LDL-c
goals despite receiving a statin alone. Bennet et al. [38],
added ezetimibe to maximally tolerated lipid lowering
therapy (statins or fenofibrate) in 33 HIV infected persons.
The mean TC and LDL-c were reduced 21% and 35%
respectively (p <0.001) and HDL-c increased 8% (p=0.038).
No adverse events occurred. Stebbin et a l. [39] analyzed
twenty nine HIV-infected persons with ezetimibe alone or
associated with statins. Of these individuals, 16 and 11
respectively were receiving PI-based and NNRTI-based
regimen. During 12 weeks, it was observed a significant
reduction of 18% in TC (p<0.01) and 28,9% in TG (p<0.05)
regardless of whether patients received statins or not, or the
type of antiretroviral therapy. In the end of study, 40% of the
patients had normalized serum TC, but the same didn’t occur
with TG. There were no significant differences in reduction
of either serum TC or TG in those receiving ezetimibe alone
(n =12) or combined with statins (n= 17). The limitations of
this study included small sample size and lack of
measurement of LDL-c level. Van den Berg-Wolf et al. [40]
in a prospective, non controlled study, evaluated twenty
HIV-infected persons who were on treatment with statins
and didn’t reach the LDL-c goal during 18 weeks. These
patients were on HAART that included ritonavir-boosted PIs
in 17 (85%) and 3 (15%) on nelfinavir. Mean percentage of
changes from baseline in LDL-c were: - 10,9%, - 12,2% and
– 9,1% at weeks 6,12,18, respectively (p<0.05 at each time
period vs baseline). No significant changes in TG and HDL-c
were seen. In a subgroup of patients on lopinavir/ritonavir
(LPV/r), the concentrations of the drug were obtained before
and after the introduction of Ezetimibe and no significant
changes were observed in the concentration of this PI.
Ezetimibe could be recommended as a second line
therapy to HAART induced dyslipidemia if hypercholesterolemia
is refractory to th e statins or if the patient does not tolerate
these drugs. Its high tolerability and the lack of interactions
with the CYP 3A4 indicate that ezetimibe will not increase
the risk of toxicity or pharmacokinetic interactions with
antiretroviral.
Fish O il
The metabolic effects of N-polyunsaturated fatty acids
(PUFAs) derived from marine sources (so-called “fish oils”)
have been demonstrated to reduce fasting and postprandial
TG levels in subjects without HIV infection [41]. Omega-3
is considered an alternative treatment in non-HIV infected
persons. Interest in the triglyceride-lowering effect of
omega-3 fatty acids first came from studies in Greenlandic
Eskimos, which showed that, despite high intake of animal
fats, there was low incidence of CAD. A contributing factor
was the type of fats being consumed, which were rich in
omega-3 fatty acids [42,43] however the exact mechanism
whereby these fats decrease the risk of CAD is unclear. It
has been demonstrated that 3-5 g per day of omega-3 fatty
acids can reduce triglycerides by 30-50%, thereby potentially
minimizing the risk of coronary heart disease (CHD) and
pancreatitis [44]. Intake of small amounts of fish has also
been shown to affect mortality [44]. One research showed
that, in addition to significantly lowering plasma levels of
TC, LDL-c and VLDL-c, salmon oil and other omega-3-
enriched fish oils can have a potentially therap eutic role in
the treatment of very high plasma TG levels [45-48]. Few
data are available on the effect of PUFAs on metabolic
abnormalities in HIV persons [49] however recent studies
suggest possible efficacy and safety of fish oil
supplementation in the treatment of antiretroviral therapy-
associated hypertriglyceridemia [50].
Whol et al. [50] conducted an open-label, randomized
trial with 52 patients receiv ing HAART with fasting TG
levels of > 200 mg/dL to receive nutritionist-administered
dietary and exercise counseling with or without fish oil
supplementation for 16 weeks. Patients assigned to receive
fish oil had a 25% decline in fasting TG levels at week 4
(95%CI, - 34.6% to - 15.7% change), compared with a 2.8%
mean increase among patients assigned to receive counseling
alone (95% CI, - 17.5% to + 23.1% change p = .007). By
Treatment of Lipid Disorders in HIV-Infected People The Open AIDS Journal, 2009, Volume 3 35
week 16, the mean reduction in TG levels in the fish oil arm
remained significant at 19.5% (95% CI, - 34.9% to – 4.0 %
change), whereas the mean decrease in the diet and exercise
only arm was 5.7% (95% CI – 24.6% to + 13.2% change);
however, the difference between study arms was no longer
statistically significant (p=.12). Low density lipoprotein
cholesterol levels had increased by 15.6% (95% CI, + 4.8%
to + 26.4 % change), at week 4 and by 22.4% (95% CI, +
7.91% to + 36.8 % change) at week 16 in the fish oil arm but
did not change in the other group. Carter et al. [51]
compared the effectiveness of omega-3 fatty acid
supplementations and placebo in lowering TG levels in HIV-
infected persons on HAART. It was a placebo-controlled,
randomized, double-blind trial in participants with stable
HAART with fasting TG > 312 mg/dL* to 890 mg/dL*
using 9 g of omega-3 fatty acids versus placebo (olive oil)
after 6 week lead in on dietary therapy. Eleven HIV-infected
males were enrolled. The mean TG level decreased from 447
mg/dL* at baseline to 395 mg/dL* (- 11.6%) after dietary
intervention and to 300 mg/dL* (-32.9%) after 8-week
treatment period. In the omega-3 fatty acid arm, TG fell from
475 mg/dL to 447mg/dL* (- 6%) after dietary intervention
and to 210 mg/dL* (- 56.9%) after treatment period. In the
placebo arm, TG fell from 425 mg/dL to 361 mg/dL*
(- 15.1%) after dietary intervention and to 363 mg/dL*
(- 14.5%) after the treatment period. The difference between
the groups was significant (p=0.0487). The estimated
difference between groups for change in mean TG over 8-
weeks was - 207 mg/dL* (CI 95% - 402 to - 11 mg/dL). De
Truchis et al. [52] evaluated the TG levels in HIV-infected
persons receiving stable antiretroviral therapy treated with
PUFAs in a prospective double-blind randomized design.
One hundred twenty two patients with TG > 200 mg/dL* and
1000 mg/dL* after 4-week diet were randomized for 8-
weeks to N-3 PUFAs (2 capsules containing 1g of fish oil 3
times daily, n=60) or placebo (1g of paraffin oil capsules,
n=62). An 8-week open-label phase of N-3 PUFAs followed.
The difference (PUFA versus placebo) in TG percent change
at week 8 was – 24.6% (range: -40.9% to – 8.4%; p =
0.0033), the median was - 25.5% in the PUFA group versus
1% in placebo group and mean TG levels at week 8 were
340 ±180 mg/dL* and 480 ± 310 mg/dL* respectively. The
TG levels were normalized in 22.4% (PUFA) versus 6.5%
(placebo) (p=0.013) with a 20% reduction in 58.6%
(PUFA) versus 33.9% (placebo) (p=0.007). Under the open-
label phase of N-3 PUFAs, the decrease in TG levels was
sustained at week 16 for patients in the PUFA group (mean
TG levels: 340 ± 170 mg/dL*, whereas a 21.2% decrease in
TG levels occurred in the placebo group (mean TG levels:
330 ± 140 mg/dL*. The median TG change at week 8 was -
43.6% (range Q1-Q3; 95% CI: - 66.5% to – 4.6%) for
patients with TG levels > 1000 mg/dL. Manfredi et al. [49]
in a prospective, open-label study, evaluated the efficacy and
safety profile of polyunsaturated ethyl esters of n-3 fatty
acids (PEEs) in the control of moderate hypertriglyceridemia
complicating antiretroviral-treated HIV disease, compared
with diet and exercise and with fibr ate. Patients with
moderate hypertriglyceridemia (200-500 mg/dL) while on
HAART, despite modified diet and increased physical
activity were selected. A total of 156 patients aged 36-62
years (97 men) were evaluated. Fifty four patients received
PEE at 1g twice daily; 53 subjects were treated with standard
doses of either: b ezafibrate (21 cases), fenofibrate (19 cases)
or gemfibrozil (13 cases); and the remaining 49 patients
continued a diet-exercise program and served as controls. An
analysis of 18 months showed that continued PEE
administration led to a significant decrease of mean TG of
5.6; 15.8, 13.3, 16; 15.3 and 11.6% after 3, 6, 9, 12, 15 and
18 months, respectively (P<0.0001 vs baseline levels), while
negligible changes occurred in serum cholesterolemia. Both
PEE and fibrate administration achieved a significant (p<
0.0001) amelioration of triglyceridemia compared with diet-
exercise only, although fibrates showed a better efficacy
profile vs PEE (p < 0.0001); these significance levels were
maintained throughout the entire 18-month observation
period. When comparing the efficacy of PEE with that of a
diet-exercise program, a significant difference was reached
at the 6th month (p=0.001) and was maintained until the 18th
month (p=0.005 – P< 0.0001). It showed that pharmaceutical
interventions for extreme hyperlipidaemia will continue to
be superior to dietary therapy alone and that omega-3 fatty
acid will be a useful additional intervention in HIV-infected
people with hypertriglyceridemia.
CONCLUSION
All HIV-infected persons should have their fasting
plasma lipid profile prior to starting HAART. These exams
should be repeated 2 to 3 months after starting or changing
antiretroviral therapy, every 2 to 3 months with the existing
of significant abnormalities and to assess response to lipid-
lowering therapy. It should be checked annually in the
absence of significant abnormalities or well-achieved targets
as a result of intervention. Efforts should be used including
lifestyle modification and lip id-lowering agents if it is
necessary, to achieve lipids goals. Special attention should
be given to drug interactions between lipid-lowering agents
and antiretroviral therapy. Neith er, lovastatin or simvastatin
should be used with PI and atorvastatin should be used with
caution. Rosuvastatin is a new statin with promising results
in HIV-infected people. Ezetimibe is the first lipid-lowering
drug that inhibits intestinal uptake of dietary and biliary
cholesterol at the brush border of the intestine and researches
showed that it could be used as monotherapy or combined
with statin. Omega-3 is considered an alternative treatment
in non-HIV infected populations and that this fish oil could
be used to treat high TG levels related to HAART.
REFERENCES
[1] Constans J, Pellegrin JL, Peuchant E, et al. Plasma lipids in HIV-
infected patients: a prospective study in 95 patients. Eur J Clin
Invest 1994; 24(6): 416-20.
[2] Grunfeld C, Kotler DP, Shigenaga JK, et al. Circulating interferon-
alpha levels and hypertriglyceridemia in the acquired
immunodeficiency syndrome. Am J Med 1991; 90(2): 154-62.
[3] Shor-Posner G, Basit A, Lu Y, et al. Hypocholesterolaemia is
associated with immune dysfunction in early human
immunodeficiency virus-1 infection. Am J Med 1993; 94(5): 515-9.
[4] Graham NM. Metabolic disorders among HIV-infected patients
treated with protease inhibitors: a review. J Acquir Immune Defic
Syndr 2000; 25 (Suppl 1): S4-11.
[5] Distler O, Cooper DA, Deckelbaum RJ, Sturley SL.
Hyperlipidemia and inhibitors of HIV protease. Curr Opin Clin
Nutr Metab Care 2001; 4(2): 99-103.
36 The Open AIDS Journal, 2009, Volume 3 da Silva and Bárbaro
[6] Barbaro G. Reviewing the cardiovascular complications of HIV
infection after the introduction of highly active antiretroviral
therapy. Curr Drug Targets Cardiovasc Haematol Disord 2005;
5(4): 337-43.
[7] Palella FJ, Delaney KM, Moorman AC, et al. Declining morbidity
and mortality among patients with advanced human
immunodeficiency virus infection. N Engl J Med 1998; 338: 853-
60.
[8] Manfredi R. Management of dyslipidemia in patients with HIV
disease. Clin Microbiol Infect 2000; 6(11): 579-84.
[9] Carpenter CC, Cooper DA, Fischl MA, et al. Antiretroviral therapy
in adults: updated recommendations of the International AIDS
Society-USA Panel. JAMA 2000; 283(3): 381-90.
[10] Currier JS. How to manage metabolic complications of HIV
therapy: what to do while we wait for answers. AIDS Read 2000;
10 (3): 162-9.
[11] Klein D, Hurley LB, Quesenberry CP, Jr, Sidney S. Do protease
inhibitors increase the risk for coronary heart disease in patients
with HIV-1 infection? J Acquir Immune Defic Syndr 2002; 30(5):
471-7.
[12] Mary-Krause M, Cotte L, Simon A, Partisani M, Costagliola D.
Clinical Epidemiology Group from the French Hospital Database.
Increased risk for myocardial infarction with duration of protease
inhibitor therapy in HIV-infected men. AIDS 2003; 17: 2479-86.
[13] Carr A, Samaras K, Burton S, et al. A syndrome of peripheral
lipodystrophy, hyperlipidaemia and insulin resistance in patients
receiving HIV protease inhibitors. AIDS 1998; 12: F51-F58.
[14] Carr A, Samaras K, Chisholm DJ, Cooper DA. Pathogenesis of
HIV-1-protease inhibitor-associated peripheral lipodystrophy,
hyperlipidaemia, and insulin resistance. Lancet 1998; 351 (9119):
1881-3.
[15] Fauvel J, Bonnet E, Ruidavets JB, et al. An interaction between
apo C-III variants and protease inhibitors contributes to high
triglyceride/low HDL levels in treated HIV patients. AID S 2001;
15 (18): 2397-406.
[16] Bonnet E, Ruidavets JB, Tuech J, et al. Apoprotein c-III and E-
containing lipoparticles are markedly increased in HIV-infected
patients treated with pro tease inhib itors: association with the
development of lipodystrophy. J Clin Endocrinol Metab 2001; 86
(1): 296-302.
[17] Caron M, Auclair M, Sterlingot H, Kornprobst M, Capeau J. Some
HIV protease inhibitors alter lamin A/C maturation and stability,
SREBP-1 nuclear localization and adipocyte differentiation. AIDS
2003; 17(17): 2437-44.
[18] Jones SP, Janneh O, Back DJ, Primohammed M. Altered
adipocytokines gene expression in murine 3T3-F442A adipocyte
treated with protease inhibitors and nucleoside reverse transcriptase
inhibitors. Antivir Ther 2003; 8: 1-11.
[19] Dubé MP, Stein JH, Aberg JA, et al. Guidelines for the evaluationa
and management of dyslipidemia in human immunodeficiency
vírus (HIV) – Infected Adutls Receiving Antiretroviral Therapy:
Recommendations of the HIV Medicine Association of the
Infectious Disease of America and the Adult AIDS Clinical Trials
Group. Clin Infect Dis 2003; 37: 613-27.
[20] Third Report of the National Cholesterol Education Program
(NCEP). Expert panel on detection, evaluation and treatment of
high blood cholesterol in adults (adult treatment panel III) final
report. Circulation 2002; 106: 3143-21.
[21] Fichtenbaum CJ, Gerber JG, Rosenkranz SL, et al. AIDS Clinical
Trials Group. Pharmacokinetic interactions between protease
inhibitors and statins in HIV seronegative volunteers: ACTG Study
A5047. AIDS 2002; 16(4): 569-77.
[22] Calza L, Manfredi R, Chiodo F. Statins and fibrates for the
treatment of hyperlipidaemia in HIV-infected patients receiving
HAART. AIDS 2003; 17(6): 851-9.
[23] Bottorff MB. Statin safety and drug interactions: clinical
implications. Am J Cardiol 2006; 97(8A): 27C-31C.
[24] Williams B, Feely J. Pharmacokinetic-pharmacodynamic drug in
interactions with HMG-CoA reductase inhibitors. Clin
Pharmacokinet 2002; 41: 434-70.
[25] Martin PD, Warwick MJ, Dane AL, et al. Metabolism, excretion,
and pharmacokinetics of rosuvastatin in healthy adult male
volunteers. Clin Ther 2003; 25(11): 2822-35.
[26] Cheng JW. Rosuvastatin in the management of hyperlipidemia.
Clin Ther 2004; 26(9): 1368-87.
[27] Bottaro EG, Caravello O, Scapellato PG, et al. Rosuvastatin for the
treatment of dyslipidemia in HIV-infected patients receiving highly
active antiretroviral therapy. Enferm Infecc Microbiol Clin 2008;
26(6): 325-29.
[28] Calza L, Manfredi R, Colangeli V, Pocaterra D, Pavoni M, Chiodo
F. Rosuvastatin, pravastatin, and atorvastatin for the treatment of
hypercholesterolaemia in HIV-infected patients receiving protease
inhibitors. Curr HIV Res 2008; 6(6): 572-8.
[29] Van der Lee M, Sankatsing R, Schippers E, et al. Pharmacokinetics
and pharmacodynamics of combined use of lopinavir/ritonavir and
rosuvastatin in HIV-infected patients. Antivir Ther 2007; 12(7):
1127-32.
[30] Merck/Schering-Plough Pharmaceuticals. Zetia (ezetimibe) product
information. North Wales, PA, 2005 .
[31] Negredo E, Molto J, Puig J, et al. Ezetimibe, a promising lipid-
lowering agent for the treatment of dyslipidaemia in HIV-infected
patients with poor response to statins. AIDS 2006; 20(17): 2159-
64.
[32] Gagné C, Bays HE, Weiss SR, et al. Ezetimibe Study Group.
Efficacy and safety of ezetimibe added to ongoing statin therapy
for treatment of patients with primary hypercholesterolemia. Am J
Cardiol 2002; 90 (10):1084-91.
[33] Pearson TA, Denke MA, McBride PE, Battisti WP, Brady WE,
Palmisano J. A community-based, randomized trial of ezetimibe
added to statin therapy to attain NCEP ATP III goals for LDL
cholesterol in hypercholesterolemic patients: the ezetimibe add-on
to statin for effectiveness (EASE) trial. Mayo Clin Proc 2005;
80(5): 587-95.
[34] Kosoglou T, Statkevich P, Johnson-Levonas AO, Paolini JF,
Bergman AJ, Alton KB. Ezetimibe: a review of its metabolism,
pharmacokinetics and drug interactions. Clin Pharmacokinet 2005;
44 (5): 467-94.
[35] Stein E, Stender S, Mata P, et al. Ezetimibe Study Group.
Achieving lipoprotein goals in patients at high risk with severe
hypercholesterolemia: efficacy and safety of ezetimibe co-
administered with atorvastatin. Am Heart J 2004; 148(3): 447-55.
[36] Farnier M, Freeman MW, Macdonell G, et al. The Ezetimibe Study
Group. Efficacy and safety of the coadministration of ezetimibe
with fenofibrate in patients with mixed hyperlipidaemia. Eur Heart
J 2005; 26(9): 897-905.
[37] Coll B, Aragonés G, Parra S, Alonso-Villaverde C, Masana L.
Ezetimibe effectively decreases LDL-cholesterol in HIV-infected
patients. AIDS 2006; 20(12): 1675-7.
[38] Bennett MT, Johns KW, Bondy GP. Ezetimibe is effective when
added to maximally tolerated lipid lowering therapy in patients
with HIV. Lipids Health Dis 2007; 6: 15.
[39] Stebbing J, Asghar AK, Holmes P, Bower M, Isenman HL, Nelson
M. Use of ezetimibe during HIV infection. J Antimicrob
Chemother 2009; 63(1): 218-20.
[40] Berg-Wolf MV, Klibanov OM, Gaughan JP, Tedaldi EM.
Ezetimibe combined with low-dose statin effectively lowers LDL
in protease inhibitor treated patients. AIDS Patient Care STDS
2008; 22(6): 483-8.
[41] Simons LA, Hickie JB, Balasubramaniam S. On the effects of
dietary n-3 fatty acids (Maxepa) on plasma lipids and lipoproteins
in patients with hyperlipidaemia. Atherosclerosis 1985; 54(1): 75-
88.
[42] Bang HO, Dyerberg J, Hjøorne N. The composition of food
consumed by Greenland Eskimos. Acta Med Scand 1976; 200(1-2):
69-73.
[43] Weber P, Raederstorff D. Triglyceride-lowering effect of omega-3
LC-polyunsaturated fatty acids - a review. Nutr Metab Cardiovasc
Dis 2000; 10(1): 28-37.
[44] O'Keefe JH Jr, Harris WS. From Inuit to implementation: omega-3
fatty acids come of age. Mayo Clin Proc 2000; 75(6): 607-14.
[45] Harris WS, Connor WE, McMurry MP. The comparative
reductions of the plasma lipids and lipoproteins by dietary
polyunsaturated fats: salmon oil versus vegetable oils. Metabolism
1983; 32(2): 179-84.
[46] Stone NJ. Fish consumption, fish oil, lipids, and coronary heart
disease. Circulation 1996; 94(9): 2337-40.
[47] Perez CF, Perales J, Fraile G. The hypolipemic effect of
concentrated salmon oil rich in n-3 fatty acids. An Med Intern a
1990; 7: 299-303.
[48] Kris-Etherton PM, Harris WS, Appel LJ. American Heart
Association. Nutrition Committee. Fish consumption, fish oil,
Treatment of Lipid Disorders in HIV-Infected People The Open AIDS Journal, 2009, Volume 3 37
omega-3 fatty acids, and cardiovascular disease. Circulation 2002;
106 (21): 2747-57.
[49] Manfredi R, Calza L, Chiodo F. Polyunsaturated ethyl esters of n-3
fatty acids in HIV-infected patients with moderate
hypertriglyceridemia: comparison with dietary and lifestyle
changes, and fibrate therapy. J Acquir Immune Defic Syndr 2004;
36 (3): 878-80.
[50] Wohl DA, Tien HC, Busby M, et al. Randomized study of the
safety and efficacy of fish oil (omega-3 fatty acid) supplementation
with dietary and exercise counseling for the treatment of
antiretroviral therapy-associated hypertriglyceridemia. Clin Infect
Dis 2005; 41(10): 1498-504.
[51] Carter VM, Woolley I, Jolley D, Nyulasi I, Mijch A, Dart A. A
randomised controlled trial of omega-3 fatty acid supplementation
for the treatment of hypertriglyceridemia in HIV-infected males on
highly active antiretroviral therapy. Sex Health 2006; 3: 287-90.
[52] De Truchis P, Kirstetter M, Perier A, et al. Reduction in
triglyceride level with N-3 polyunsaturated fatty acids in HIV-
infected patients taking potent antiretroviral therapy: a randomized
prospective study. J Acquir Immune Defic Syndr 2007; 44(3): 278-
85.
[53] Calza L, Manfredi R, Pocaterra D, Chiodo F. Risk of premature
atherosclerosis and ischemic heart disease associated with HIV
infection and antiretroviral therapy. J Infect 2008; 57(1): 16-32.
[54] Soler A, Deig E, Guil J, Rodríguez-Martín M, Guelar A, Pedrol E.
Effectiveness and tolerance of atorvastatin for antiretroviral
therapy-secondary dyslipemia. Med Clin (Barc) 2006; 127(7): 250-
2.
[55] Bader MS, Kelly DV. Diagnosis and management of common
chronic metabolic complications in HIV-infected patients. Postgrad
Med 2008; 120(4): 1-11.
Received: March 11, 2009 Revised: May 15, 2009 Accepted: May 19, 2009
© da Silva and Bárbaro; Licensee Bentham Open.
This is an open access article licen sed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-
nc/3.0/) which permits unrestricted, non-commercial use, distribution and reproduction in any medium, provided the work is properly cited.
... Since the introduction of highly active antiretroviral therapy (HAART), the life expectancy of HIV patients has extended notably, raising concerns about the impact of prolonged exposition to metabolic disorders on their long-term prognosis and outcome [14]. ...
... In all of the treatment schemes, ezetimibe was found to be effective, safe and well tolerated, due to the lack of influence on CYT P450, and it presented a lack of interactions while showing no risk of increasing the antiretroviral toxicity [4,14,47,70]. In coadministration with other lipid lowering agents [4,14,47,70] as well as in monotherapy, [10,70,78] ezetimibe was efficient in diminishing LDL levels. ...
... In all of the treatment schemes, ezetimibe was found to be effective, safe and well tolerated, due to the lack of influence on CYT P450, and it presented a lack of interactions while showing no risk of increasing the antiretroviral toxicity [4,14,47,70]. In coadministration with other lipid lowering agents [4,14,47,70] as well as in monotherapy, [10,70,78] ezetimibe was efficient in diminishing LDL levels. ...
Article
Ezetimibe is the first agent used in hypercholesterolemia treatment known to lower intestinal cholesterol uptake that is able to inhibit NPC1L1 transport proteins in the brush boarder of enterocytes and macrophages. Furthermore, it demonstrates anti-inflammatory and immunomodulatory properties and influences the expression of certain antigens. The drug is rapidly absorbed from the gastrointestinal tract and is then glucuronidated to form the active metabolite. It also undergoes extensive enterohepatic circulation. Various genetic polymorphisms seem to influence the pharmacokinetics of ezetimibe with different effects. The drug also presents a complex impact on cytochrome P450 enzymes, as it is a metabolism-dependent inhibitor of CYP3A4. Ezetimibe does not demonstrate any clinically significant interactions with statins, fibrates, mipomersen sodium, levothyroxine or lopinavir. However, its effect in conjunction with cyclosporine is not neutral. The use of this cholesterol absorption inhibitor has been shown to be safe and effective among patients after cardiac, renal and liver transplants, as well as in HIV patients.
... Publications about dyslipidemia management for HIV mention fish oil (supplement of EPA/DHA) as a safe and alternative treatment to reduce serum TG because it is well tolerated and does not present drug interactions with antiretroviral therapy [37][38][39] .Recommended dosage vary from 2 to 9g/d and the supplements are indicated some times as fish oil, others as Omega 3 fatty acids and others as EPA/DHA 37,38,[40][41][42] . ...
Article
Full-text available
Hypertriglyceridemia is common in antiretroviral therapy-treated patients and Omega 3 fatty acids are being used as a intervention in reducing serum triglycerides (TG) in these patients. The objective of this study is to evaluate the effectiveness of the use of Omega 3 in the treatment of hypertriglyceridemia in HIV/AIDS patients on antiretroviral therapy. This study is a systematic review with meta-analysis of randomized clinical trials. Electronic databases – PubMed, Cochrane and Lilacs were researched. Fifty one articles were encountered. Nine were added to the meta-analysis. The reduction of triglycerides level was -77.55 mg (IC of -121.85 to -33.25) in Omega 3 groups. The analysis considering trials with more than 1000 mg of EPA/DHA included seven studies and the heterogeneity dropped to 0%.The reduction of combined averages was -101.56mg (IC of -145.76 to -57.37). The analysis considering trials with patients that had more than 200 mg/dL of initial triglycerides included also seven trials and the heterogeneity dropped to 0%. The reduction of combined averages was -114.15 mg (IC of -162.34 to -65.97). EPA/DHA supplementation reduces serum triglycerides levels in patients with HIV/AIDS-associated hypertriglyceridemia in stable use of antiretroviral therapy.
... 7,8,10 Hyperlipidemia and severe isolated hypertriglyceridemia (triglyceride level > 1000 mg/dL) require intervention to reduce the risk of cardiovascular events and pancreatitis, respectively, in persons living with HIV. 8,[11][12][13] The fibric acid derivatives, gemfibrozil and fenofibrate, are commonly used for the management of severe or isolated hypertriglyceridemia. 14 In a retrospective cohort study, over 100 hypertriglyceridemic, HIV-infected patients who were treated with gemfibrozil and also receiving protease inhibitor-based cART, had a smaller mean change in triglyceride levels (−44.0%) compared with HIV-infected patients who received nonnucleoside reverse transcriptase inhibitor (NNRTI)-based cART (-60.3%) or non-HIV-infected controls (-59.3%). ...
Article
Study Objective Because we previously observed a significant 41% reduction in gemfibrozil exposure after 2weeks of lopinavir-ritonavir administration, we sought to determine the influence of lopinavir-ritonavir and ritonavir alone on the pharmacokinetics of fenofibric acid, an alternative to gemfibrozil for the treatment of elevated triglyceride levels. DesignOpen-label, single-sequence pharmacokinetic study. SettingClinical Research Center at the National Institutes of Health. SubjectsThirteen healthy adult volunteers. InterventionSubjects received a single oral dose of fenofibrate 145mg during three study phases: before ritonavir administration, after 2weeks of administration of ritonavir 100mg twice/day, and after 2weeks of administration of lopinavir 400mg-ritonavir 100mg twice/day. Measurements and Main ResultsSerial blood samples were collected over 120hours for determination of fenofibric acid concentrations. Fenofibric acid pharmacokinetic parameter values were compared before and after concomitant ritonavir or lopinavir-ritonavir administration. The geometric mean ratios (90% confidence intervals) for fenofibric acid area under the plasma concentration-time curve were 0.89 (0.77-1.01) after 14days of ritonavir alone compared with baseline (p>0.05) and 0.87 (0.69-1.05) after 14days of lopinavir-ritonavir compared with baseline (p>0.05). Study drugs were generally well tolerated; all adverse events were mild or moderate, transient, and resolved without intervention. Conclusion In contrast to a significant interaction between gemfibrozil and lopinavir-ritonavir, neither lopinavir-ritonavir nor ritonavir alone altered the pharmacokinetics of fenofibric acid in healthy volunteers. These data suggest that fenofibrate remains an important option in human immunodeficiency virus-infected patients receiving common ritonavir-boosted therapy.
... The search for vaccines to protect against HIV/AID has continued to elude the science world. However, the use of highly active antiretroviral therapy (HAART) has dramatically improved the prognosis of HIV-infected individuals 1,2 .The beneficial effect of reduced risk of early death from opportunistic infections and other consequences of HIV infection, is however, reduced because other possible causes of death have replaced this beneficial effect 3 . Myocardial infarction has become a matter of particular concern. ...
Article
Background: Diet and genetic predisposition significantly affect lipid metabolism in the individual. This metabolic effect is further challenged in patients infected with HIV and on HAART. The prolonged use of HAART is associated with lipodystrophy, dyslipidemia, and insulin resistance. Objective: To determine the prevalence of lipid dysregulation and dysglycaemia in HIV infected patients on HAART in the Kumasi metropolis. Methods: This cross sectional study was conducted between October 2009 and June 2010, and 305 HIV-infected patients consisting of 164 patients on HAART for at least six months and 141 HAART-naive patients constituted HIV-positive patients, not on HAART and whose CD4 were not below 320 cell/ml as the control. Data was analyzed using Graph Pad Prism (version 5.0). Unpaired t-test, linear and multivariate regression analyses, was used to predict glucose level from the various parameters. Anthropometric parameters consisting of body weight, waist and hip circumferences, height, bicep and triceps skin fold were measured with a pair of calipers. Lipid profile and fasting blood glucose were determined by enzymatic methods. CD4 counts and hemoglobin were determined. Results: Fasting plasma, glucose (3.81±0.08mmol/l, 4.48±0.17mmol/l), total cholesterol (3.05± 0.0 8mmol/l, 4.54±0.08mmol/l) LDL (2.24±0.07mmol/l, 2.87±0.07mmol/l) and HDL (0.85±0.04mmol/l, 0.97±0.03mmol/l) between the control and case respectively were significantly raised (P< 0.001), though within the physiological range. The significantly increased hip and waist circumferences, waist-to-hip ratio (0.85±0.22, 0.88±0.01) of the control and case correlated with lipodystrophy. Conclusion: HAART was associated with lipodystrohy and, the risk of developing type II diabetes among the HAART experienced group was 5 times higher than the HAART naive group.
Article
Full-text available
Background: Premature atherosclerosis in HIV-infected patients is associated with chronic infection by itself and adverse effects of antiretroviral treatment (ART). Extra virgin olive oil (EVOO) has a beneficial effect on the cardiovascular system because of its anti-inflammatory properties. The objective of this study was to determine whether the consumption of EVOO improves inflammation and atherosclerosis biomarkers in HIV-infected patients receiving ART. Material/Methods: This randomized, crossover, controlled trial included 39 HIV-positive male participants who consumed 50 mL of EVOO or refined olive oil (ROO) daily. Four participants dropped out of the study. Leukocyte count, erythrocyte sedimentation rate (ESR), high-sensitivity C-reactive protein (hsCRP), interleukin-6, fibrinogen, total cholesterol, LDL cholesterol, HDL cholesterol, triglycerides, malondialdehyde, glutathione-peroxidase, superoxide dismutase, oxidized LDL and von Willebrand factor were determined before the first and after each of the 2 intervention periods. Intervention and washout periods lasted for 20 and 14 days, respectively. Results: In participants with >90% compliance (N=30), hsCRP concentrations were lower after EVOO intervention (geometric mean [GM], 1.70 mg/L; 95% confidence interval [CI], 1.15–2.52) compared to ROO administration (GM, 2.92 mg/L; 95% CI, 1.95–4.37) (p=0.035). In participants using lopinavir/ritonavir, ESR and hsCRP concentrations decreased 62% and 151%, respectively, after EVOO administration. In the whole study population (N=35) we found no difference in analyzed biomarkers after EVOO administration. Conclusions: Our exploratory study suggests that EVOO consumption could lower hsCRP in patients on ART.
Article
Full-text available
HIV dan AIDS telah menjadi pandemi. Lebih dari 30 juta orang terinfeksi HIV terutama di daerah Afrika-Sahara. Penyakit ini dahulu memiliki tingkat kematian tinggi, namun semenjak digunakannya Highly Active Anti Retroviral Therapy (HAART), mortalitas penyakit ini menurun dengan drastis, bahkan mengubah status AIDS menjadi penyakit kronis layaknya hipertensi dan diabetes mellitus. Penurunan morbiditas dan mortalitas HIV dan AIDS meningkatkan kualitas hidup Orang Dengan HIV dan AIDS (ODHA) secara siginifikan, namun di samping itu, efek simpang jangka panjang anti retroviral (ARV) bermunculan, di antaranya terkait dengan sistem kardiovaskuler. Lipodistrofi salah satu keadaan akibat komplikasi metabolik dan distribusi lemak yang terganggu banyak ditemui pada pasien dengan terapi kombinasi analog nukleosida dan protease inhibitor (PI). Prevalensi lipodistrofi antara 30-50%. Tetapi menurut beberapa penelitian, hubungan antara PI dan lipodistrofi masih belum jelas. Perlu dilakukan penggantian NRTI atau PI yang menyebabkan lipodistrofi.
Article
Full-text available
HIV and AIDS has become a pandemic. More than 30 millions are HIV infected particularly in Sub-Sahara Region. Once the disease is known for its high mortality rate, however, since the use of Highly Active Anti Retroviral Therapy (HAART), the mortality has been decreased substantially. Moreover, HAART has changed the status of AIDS to become a chronic disease like hypertension and diabetes mellitus. The decrease of morbidity and mortality of HIV and AIDS that leads to increase the quality of life of the people living with HIV and AIDS (PLWHA) significantly. Though the adverse events on cardiovascular system that emerges from the long-term use of ARVs has been noticed. Lipodystrophy is one of the notorious metabolic complications and altered fat distribution which mostly found among patients treated by combination therapy of nucleoside analogues and protease inhibitors (PIs). The prevalence of lipodystrophy is between 30-50%. But, according to many studies, the relationship between PIs and lipodystrophy is not clear yet. It is advised to switch to "non-lipodistrophy-inducing" ARVs.
Article
Antiretroviral therapy improved the survival of patients with human immunodeficiency virus (HIV) infection. With increased life expectancy, HIV-infected patients increasingly are experiencing comorbidities, most notably cardiovascular risk factors (CRFs) and coronary heart disease (CHD). This study utilized a prospective, cross-sectional multicentre long-term design. In 803 patients (82% male; mean age 44.2 ± 10.3 years) we evaluated the prevalence of CRFs and 10-year risk of CHD using the Framingham risk model. The presence of a risk factor was determined based on the guidelines of the National Cholesterol Education Program (NCEP ATP III), the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure (JNC7), and the German Society of Cardiology. The most common CRFs were smoking (51.2%), high triglycerides (39.0%), low high-density lipoprotein cholesterol (27.5 %) and high blood pressure (21.4%). In total, 60.3%, 21.6%, and 18.1% of patients were categorized as being at low (<10%), moderate (10-20%), and high (>20%) 10-year risk for CHD, respectively. In patients with hypertension, at least one antihypertensive drug was given in 91/163 (55.8%) patients. The percentage of patients on treatment with diabetes mellitus was 23/41 (56.1%). Anti-platelet therapy was prescribed to 42/102 (41.2%) patients with known CHD or CHD equivalent. In patients of moderate or high CHD risk there were more than 50% and 30% for LDL cholesterol and more than 60% and 40% for total cholesterol untreated. The prevalence of CRFs remains high in an HIV-infected population. CRF management of HIV-infected patients deserves further improvement.
Article
Full-text available
Background and objective We investigated atorvastatin effectiveness and tolerance in HIV patients with hypercholesterolemia related to antiretroviral treatment. Patients and method Prospective study that included HIV+ patients under antiretroviral treatment who displayed secondary dyslipemia and medical treatment criteria (according to NCEP-III). These patients were given 10 mg/day atorvastatin and hygienic-dietetic measures. If the therapeutic objectives were not achieved, the dose of atorvastatin was increased to 20 mg/day. Patients were followed up for 6 months. Results 32 patients were included. In 5 cases it was necessary to increase the dose from 10 mg atorvastatin to 20 mg. The therapeutic objective was obtained in 62% cases, with a good clinical tolerance. Only one adverse effect was noticed, which forced the removal of the drug. Conclusion In our study atorvastatin was effective for the treatment of dyslipemia in HIV patients, and it was safe and well tolerated.
Article
Long-term therapy with protease inhibitors (PIs) can induce hypertriglyceridemia and development of a lipodystrophy. To better understand these metabolic alterations, the apoprotein and lipoparticle profile was investigated in male HIV patients under antiretroviral therapy: 49 received PIs, and 14 were given only two reverse transcriptase inhibitors. As controls, 63 male subjects were selected from a population study carried out in the Toulouse, France, area. Fasting glucose, insulin, and C-peptide were also determined. All patients under PIs displayed low levels of plasma glucose and increased insulin. PI administration was associated with moderate hypertriglyceridemia, low high-density cholesterol and apolipoprotein (apo) A-I levels. The most striking changes were a 2- to 3-fold increase in apo E and apo C-III, essentially recovered as associated to apo B-containing lipoparticles. Levels of those lipoparticles were two to eight times above control values. About 50% of PI-treated patients had developed a patent lipodystrophy. Multivariate analysis revealed that, among the investigated parameters, apo C-III was the only one found strongly associated with the occurrence of lipodystrophy (odds ratio, 5.5; P < 0.015). Finally, 13 PI-receiving subjects with patent hypertriglyceridemia were given fenofibrate and were reevaluated 2 months later. Triglycerides, apo E, apo C-III, and the corresponding lipoparticles had returned to nearly normal levels. These results document the accumulation of potentially atherogenic lipoparticles under PIs. Apo C-III may play a pivotal role in the development of hypertriglyceridemia and lipodystrophy.
Article
Currently available protease inhibitors are associated with development of a group of metabolic disorders. These include a peripheral lipodystrophy syndrome in which there is fat wasting in the face, arms, and legs; fat accumulation in the abdomen, dorsocervical region, and/or breasts (women only); as well as hyperlipidemia. hypercholesterolemia. and insulin resistance. A review of 15 observational studies and case reports shows that the incidence of the peripheral lipodystrophy syndrome increases with time of exposure to protease inhibitors, with a >60% incidence seen after 1 year of continuous treatment. Protease inhibitors are hypothesized to cause this syndrome by impairing conversion of retinoic acid to cis-9-retinoic acid (leading to impaired peripheral fat storage, sequestration of body fat to central adipocytes, and hyperlipidemia) and by inhibiting low-density lipoprotein receptor-related protein (LRP), thus preventing postprandial chylomicron clearance and further contributing to hyperlipidemia. Recent in vitro data suggest that more than one pathway contributes to the lipodystrophy syndrome and that pathways may differ among protease inhibitors. Although the central fat accumulation, hyperlipidemia, and insulin resistance components of this syndrome may reverse after discontinuation of protease inhibitor therapy, it is not known whether complete normalization of fast-wasted body regions is possible. Prospective controlled studies are needed to define whether protease inhibitors currently under development are less prone to produce the lipodystrophy syndrome.
Article
Objective: To describe a syndrome of peripheral lipodystrophy (fat wasting of the face, limbs and upper trunk), hyperlipidaemia and insulin resistance in patients receiving potent HIV protease inhibitor therapy. Design: Cross-sectional study. Setting: Outpatient clinic of a university teaching hospital. Patients: HIV-infected patients either receiving at least one protease inhibitor (n = 116) or protease inhibitor-naive (n = 32), and healthy men (n = 47). Interventions and main outcome measures: Lipodystrophy was assessed by physical examination and questionnaire and body composition by dual-energy X-ray absorptiometry. Fasting triglyceride, cholesterol, free fatty acid, glucose, insulin, C-peptide and fructosamine levels, other metabolic parameters, CD4 lymphocyte counts, and HIV RNA load were also assessed. Results: HIV protease inhibitor-naive patients had similar body composition to healthy men. HIV protease inhibitor therapy was associated with substantially lower total body fat (13.2 versus 18.7 kg in protease inhibitor-naive patients; P = 0.005), and significantly higher total cholesterol and triglyceride levels. Lipodystrophy was observed clinically in 74 (64%) protease inhibitor recipients after a mean 13.9 months and 1(3%) protease inhibitor-naive patient (P = 0.0001). Fat loss occurred in all regions except the abdomen after a median 10 months. Patients with lipodystrophy experienced a relative weight loss of 0.5 kg per month and had significantly higher triglyceride, cholesterol, insulin and C-peptide levels and were more insulin-resistant than protease inhibitor recipients without lipodystrophy. Patients receiving ritonavir and saquinavir in combination had significantly lower body fat, higher lipids and shorter time to lipodystrophy than patients receiving indinavir. Three (2%) patients developed new or worsening diabetes mellitus. Conclusion: A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance is a common complication of HIV protease inhibitors. Diabetes mellitus is relatively uncommon.
Article
Background: In the current context of dyslipidaema, hyperglycaema and lipodystrophia observed among HIV-seropositive subjects, it is important to study the risk of myocardial infarction (MI) in this population. The French Hospital Database on HIV, which includes a large number of seropositive subjects followed for substantial periods, offers the opportunity to analyse the impact of protease inhibitors (PI) on the risk of MI among men. Methods: Cox model was used to study the risk factors of MI occurrence. Standardized morbidity ratios (SMR) in men exposed to PI were calculated with data from the French general male population (FGMP) of the same age as reference. Results: Between 1996 and 1999, MI was diagnosed in 60 men among 88 029 person-years (PY), including 49 cases among men exposed to PI. In the Cox model, exposure to PI was associated with a higher risk of MI [relative hazard (RH), 2.56; 95% confidence interval (CI), 1.03-6.34]. The expected incidence in the FGMP was 10.8/10 000 PY. The SMR relative to the FGMP was 0.8 (95% CI, 0.5-1.3) for men exposed to PI for < 18 months (G1), 1.5 (95% CI, 0.8-2.5) for men exposed for 18-29 months (G2) and 2.9 (95% CI, 1.5-5.0) for men exposed for ≥ 30 months (G3). With G1 as reference, the SMR was 1.9 (95% CI, 1.0-3.1) for G2 and 3.6 (95% CI, 1.8-6.2) for G3. Conclusion: Our results point to a duration-related effect relationship between PI and MI, with a higher MI incidence rate among men exposed to PI for 18 months or more.
Article
Purpose: Patients with the acquired immunodeficiency syndrome exhibit marked disturbances in lipid metabolism. Because altered lipid metabolism may affect immune processes, this study characterized the lipid profile of asymptomatic individuals infected with the human immunodeficiency virus (HIV-1), in relationship to immune function. Patients and methods: Serum levels of triglycerides and cholesterol were determined in 94 asymptomatic HIV-1-infected (Centers for Disease Control stage II, III) homosexual men and 42 healthy seronegative control subjects. Immune assessment included measurements of lymphocyte subpopulations (CD4), immune activation (beta 2-microglobulin), natural killer cell function, and lymphocyte proliferation in response to mitogens phytohemagglutinin and pokeweed. Dietary intake was determined using a semiquantitative food frequency questionnaire. Results: Despite greater consumption of saturated fat and cholesterol, significantly lower levels of total, high-density, and low-density lipoprotein cholesterol were observed in HIV-1-seropositive men, relative to seronegative controls (p < 0.05), with 40% of the HIV-1-infected group demonstrating hypocholesterolemia (less than 150 mg/dL). Low values of total, high-density, and low-density cholesterol were associated with elevated levels of beta 2-microglobulin in HIV-1-seropositive men. No difference between the groups was noted for serum triglycerides. HIV-1-infected subjects did not demonstrate the significant inverse relationship between cholesterol and mitogen response observed in seronegative controls. Conclusions: These findings indicate that low levels of cholesterol are prevalent during the early stages of HIV-1 infection and associated with specific alterations in immune function, suggesting that hypocholesterolemia may be a useful marker of disease progression.